Skip to main content
International Journal of Alzheimer's Disease logoLink to International Journal of Alzheimer's Disease
. 2010 Oct 5;2010:859101. doi: 10.4061/2010/859101

Gene-Environment Interaction Research and Transgenic Mouse Models of Alzheimer's Disease

L Chouliaras 1, A S R Sierksma 1, G Kenis 1, J Prickaerts 1, M A M Lemmens 1, I Brasnjevic 1, E L van Donkelaar 1, P Martinez-Martinez 1, M Losen 1, M H De Baets 1, N Kholod 1, F van Leeuwen 1, P R Hof 2, J van Os 1, 3,3, H W M Steinbusch 1, D L A van den Hove 1, 4,4, B P F Rutten 1,*
PMCID: PMC2952897  PMID: 20953364

Abstract

The etiology of the sporadic form of Alzheimer's disease (AD) remains largely unknown. Recent evidence has suggested that gene-environment interactions (GxE) may play a crucial role in its development and progression. Whereas various susceptibility loci have been identified, like the apolipoprotein E4 allele, these cannot fully explain the increasing prevalence of AD observed with aging. In addition to such genetic risk factors, various environmental factors have been proposed to alter the risk of developing AD as well as to affect the rate of cognitive decline in AD patients. Nevertheless, aside from the independent effects of genetic and environmental risk factors, their synergistic participation in increasing the risk of developing AD has been sparsely investigated, even though evidence points towards such a direction. Advances in the genetic manipulation of mice, modeling various aspects of the AD pathology, have provided an excellent tool to dissect the effects of genes, environment, and their interactions. In this paper we present several environmental factors implicated in the etiology of AD that have been tested in transgenic animal models of the disease. The focus lies on the concept of GxE and its importance in a multifactorial disease like AD. Additionally, possible mediating mechanisms and future challenges are discussed.

1. Introduction

Alzheimer's disease (AD) is the most common form of dementia, characterized by an initial loss of short-term memory, followed by a progressive impairment in multiple cognitive domains. The estimated lifetime risk for developing AD is about 20% for women and 10% for men aged above 65 [1]. The pathology of AD is characterized by an accumulation of misfolded proteins, oxidative damage, and inflammatory changes ultimately resulting in region-specific loss of synaptic contacts and neuronal cell death [2]. Current biological theories on the etiology and pathology of AD posit central roles for age-related molecular and cellular aberrations that induce an imbalance in the production, cleavage, and clearance of amyloid-β (Aβ), hyperphosphorylation of the tau protein, and aberrant apolipoprotein E (APOE) function in the aging brain [1]. Several genetic risk factors have been linked with an increased risk of developing AD, such as mutations in the amyloid precursor protein (APP) and presenilin (PS) 1 and 2 for the familial cases of AD (FAD), as well as the APOE4 allele for the sporadic late-onset form of AD (LOAD). Several new genetic findings derived from powerful genome-wide association studies (GWAS; see below) have confirmed that AD is a polygenic disorder. The genes identified in these studies may enlighten unknown biological pathways involved in AD [3].

Furthermore, various environmental exposures have been found to modify the risk of AD, such as diet and nutrition, physical exercise, exposure to metals, and brain trauma. Comorbidities, such as vascular disorders or depression, could also be of considerable importance, since these have also been suggested to contribute to the risk of AD. Recent evidence indicates that more attention should be paid to the role of the environment and its interactions with underlying genetic susceptibility in triggering disease-related phenotypes [4]. The gene-environment interaction (GxE) approach differs from the linear approach of either genetic or environmental effects by positing a causal role not only for either genes or environmental exposures in isolation, but for their synergistic participation in leading to a certain phenotype (here AD), where the effect of one is conditional for the other [57]. Where epidemiological studies on AD may reveal statistical evidence for GxE in the onset and course of AD, animal research can be instrumental in studying the underlying biological mechanisms.

1.1. Objective

The objective of this review is to give an overview of the available transgenic mouse studies on AD, specifically addressing the concept of GxE. We start with a brief description of the various genetic and environmental risk factors of AD, and the different available transgenic mouse models of AD. The main part of the paper describes the effects of several environmental exposures on AD-related phenotypes. These sections begin with a brief description of the epidemiological evidence in AD (when available from meta-analyses) and continue with describing the findings from experimental animal studies in which the environmental factor was manipulated in AD transgenic mice and, when performed, in wild-type (WT) mice. Thereafter, we discuss the strengths and limitations of these studies, and we end with identifying future challenges and prospects.

2. Alzheimer's Disease

2.1. Genetics of AD

Twin studies on AD have shown a heritability of 60%–80% and a concordance of 18% up to 83%, depending on for example, the population and age of the subjects investigated. Thus, both heritable and nonheritable factors play an important role in AD's age of onset, risk and etiology [811]. Several genetic risk factors have been linked to AD. Mutations in APP, PS1, and PS2 genes have consistently been associated with early-onset FAD. Also for LOAD several susceptibility loci have been linked with risk for AD, such as the gene encoding for the APOE4 allele or loci in the clusterin (CLU), phosphatidylinositol binding clathrin assembly protein (PICALM), complement receptor 1 (CR1), BIN1 (bridging integrator, amphiphysin) genes, a locus near the BLOC1S3 (biogenesis of lysosomal organelles complex1, subunit 3), and MARK4 (microtubule affinity-regulating kinase 4) genes [3, 1214]. Other susceptibility loci have also been associated with AD (see [12, 15], http://www.alzgene.org/).

2.2. Environment and AD

Although a range of environmental exposures have been linked to AD, well-replicated and meta-analyses' evidence for the involvement of clear environmental factors in AD is sparse. Recent studies, however, have shown that dietary factors, such as exposure to a Mediterranean diet, fish and high omega-3 diets, cigarette smoking, head trauma, infections, systemic inflammation, and metal and pesticide exposure can significantly alter an individual's risk of developing AD. In addition, psychosocial factors such as education, social network, leisure activities and physical activity, chronic stress, and depression also seem to be connected to the risk of developing AD [1618]. Somatic factors that are under the direct influence of environmental exposures, such as blood pressure, obesity, diabetes mellitus, cardio- and cerebrovascular diseases, and hyperlipidemia, have additionally been implicated in AD etiology [16, 18].

2.3. Gene-Environment Interactions and AD

The field of GxE research appears very promising for psychiatry and neuroscience, albeit still little investigated in AD [19]. The notion of potential existence of GxE in AD has substantial impact on the interpretation of reports on genetic and nongenetic contribution to this disorder. Reported contributions of environmental and genetic factors to disease risk can be misleading, since they represent the environmental exposure in relationship with the genetic susceptibility or resilience to it [6]. Thus, the advantage of the concept of GxE is that it includes the genetic control of sensitivity to the environment. Additionally, the genome-wide genetic findings identify associations that also include underlying GxE [6]. In fact, evidence for GxE in AD has recently started to accumulate. For example, an interaction between the APOE4 allele and cholesterol levels has been shown to increase the risk of AD [20, 21]. Significant statistical interactions were also found between moderate consumption of alcohol and the APOE4 genotype, as well as for smoking and the APOE4 genotype [22, 23]. Furthermore, an interaction with this risk genotype and social factors, such as cohabiting with a partner has been found; APOE4 carriers who lost their partner before midlife showed an increased risk of developing AD, compared to married or cohabiting people [24].

These epidemiological studies indicate that it makes sense to focus future clinical AD studies on measuring both genes and environment and analyzing possible interactions, given that certain environmental factors may only affect a phenotype when the person is genetically endowed. A major drawback of epidemiological clinical studies is that they may indicate merely statistical interactions and thus cannot easily decipher the biological mechanisms that underlie the observed statistical interactions. Other major obstacles in clinical studies are the heterogeneity of the study population and co-occurrence of various environmental exposures in the same individuals. Experimental animal research has the advantage of enabling strict control of genetic and environmental variables. Recent advances in transgenesis allow altering specific genes in isolation, and in a time- and region-specific manner. As such, transgenic mice form a useful tool to study the effects of genetic and environmental variations and to identify the biological mechanisms that underlie the statistical GxE interactions observed in epidemiological studies (see Figure 1).

Figure 1.

Figure 1

Research in Alzheimer's disease (AD) uses both clinical (human) and preclinical (mouse) methods to elucidate the underlying mechanisms of AD etiology. Epidemiological findings such as genetic and environmental risk factors can provide tools for investigating their effects on AD etiology separately in mouse models of AD. In this paper it is, however, postulated that AD research should move towards a gene-environment (GxE) interaction approach, so that the synergistic participation of genes and environment can be scrutinized. Genes in the dashed box represent those genes found to be implicated with Alzheimer's disease etiology in humans, while genes in the solid box resemble the genes that are currently used in mouse models of Alzheimer's disease. APOE4: Apolipoprotein ε4; APP: amyloid precursor protein; CLU: clusterin; EMF: electromagnetic field; PICALM: phosphatidylinositol-binding clathrin assembly protein; PS1: Presenilin 1; PS2: Presenilin 2.

2.4. Transgenic Mouse Models of AD

Without the intention of giving a full overview of the available AD mouse models, some details on the types of transgenic mice that are discussed in the present paper can be found in Table 1. Information on the promoters used for the transgenic construct, and further details on genetic background are not further discussed here as these aspects lie outside the scope of this paper.

Table 1.

Transgenic mouse models of Alzheimer's disease with reported environmental effects.

Name Mutation Background Effect Ref.
3xTg Injection of APPswe and tauP301L transgenes in PS1M146V knock-in mice 129/C57BL6 Intraneuronal Aβ at 3 months, extracellular at 6, hippocampal hyperphosphorylated tau pathology at 12 months, synaptic dysfunction [26]

Aβ PPswe Carrying the mutant AβPPK670N, M671L gene Mixed background of 56.25% C57, 12.5% B6, 18.75% SJL, and 12.5% Swiss-Webster Amyloid deposition and cognitive decline starting at the age of 8 months [27]

APOE3, APOE4 APOE knockout mice carry an inactivated APOE endogenous gene disrupted by gene targeting in embryonic stem cells. Human APOE genomic DNA fragments injected in single cell emryos fertilized by APOE knockout mice APOE knockout and C57BL6 Expression of human APOE in the brain, high cholesterol levels [28]

APP23 cDNA of human APP with the Swedish double mutation at positions 670/671 combined with the V717I mutation, inserted to the blunt ended XhoI site of the expression cassette containing the murine Thy 1.2 gene C57BL6 Aβ deposition in the neocortex and hippocampus at the age of 6 months [29]

APP715SL Swedish (KM670/671NL) and London (V717I) mutation under control of Thy1 promotor CBA/C57BL6 Amyloid plaque deposits at 6 months of age [30]

APPNLh/NLh Human Aβ coding sequence knocked-in to the endogenous APP gene, combined with the Swedish (K670N/M671L) mutation 129/Sv No Aβ depositions, but a 9-fold increase in human Aβ production compared to normal human Aβ levels [31]

APP/PS1 KI Double knock-in mouse: APPNLh/NLh crossed with PS1 P264L knock-in, using Cre-lox knock-in technology and endogenous promoters CD-1/129 Increase of Aβ42 levels, amyloid deposition and reactive gliosis by 4 months of age. [32, 33]

APPswe/ind Expressing human APP with Swedish mutation (K670N/M671L) and the V717I Indiana mutation under the PDGF promoter (J20 line) C57BL6 × DBA/2 Increased Aβ production and Aβ deposition at 5–7 months of age, decrease in synaptophysin immunoreactivity at 2–4 months of age [34]

APPswe/PS1ΔE9 Cross of APPswe and PS1ΔE9 (expressing human PS1 carrying the exon 9 deleted variant) C57BL6J Amyloid plaque deposition, cholinergic marker decrease, memory deficits at 6 months of age [35]

APPswe/PS1Leu235Pro APP swedish mutation crossed with mutant human PS1 Leu235Pro C3H/HeJ/C57BL/A2G [36, 37]

APPswe/PS1M146L Tg2576 combined with PS1 (M146L) mutation (under PDGF promoter) C57/B6/SJL/Swiss Webster Compared to Tg2576, 41% increase in Aβ42 which precedes fibrillar Aβ deposits in cerebral cortex and hippocampus. Reduced spontaneous alternation performance in the Y-maze. [38]

APPV717I-C100 Expressing the C-terminal 100 amino acid of human APP with 717 London mutation C57BL6 Intracellular accumulation of soluble Aβ [39]
APP-YAC The entire human APP gene inserted to the yeast artificial chromosome (YAC) B142F9, introduced to embryonic stem cell by lipofection C57BL6 Significant human APP expression in the cerebral cortex [40]

PDAPP Indiana mutation (V717F) with portions of APP introns 6–8, driven by the PDGF promoter Extracellular Aβ deposits in the hippocampus from the age of 6 months and neocortex from 8 months of age [41]

PS1-L286V Overexpressing human PS1 with L286 mutation under the control of human PDGF-β promoter FVB/N Aβ42 intracellular deposits at 13 months of age [42]

TASTPM Carrying human APPswe and PS1 M146V mutations C57BL63H Cerebral Aβ deposition and cognitive deficits at 6 months of age [43]

Tg19959 TgCRND8 mice plus M146L + L286V PS1 transgene in the hamster PrP gene promoter C57/C3H/129SvEv/ Tac/FVB Amyloid deposits at 1 month of age [44]

Tg2576 Human APPswe (double K670N, M671L) inserted to hamster prion protein promoter (PrP) (is also known and referred to in the text as APPswe) C57BL6 5-fold increase in Aβ40 and 14 fold increase in Aβ42, behavioral deficits, amyloid plaques at 9 months of age [45]

TgC100 Expressing the C-terminal 100 amino acid of human APP (with or without 717 London mutation) C57BL6 Intracellular accumulation of soluble Aβ [39]

TgCRND8 Swedish and Indiana (V717F) APP mutations C57/C3H/129SvEv/ Tac/FVB Plaques at 3 months of age, increased Aβ42/40 ratio [46]

TgV337M V337M longest tau, cDNA inserted to the PDGFβ-chain expression vector B6SJL Hyperphosphorylated tau aggregates in the hippocampus, neurodegeneration, reduced hippocampal neural activity and behavioral abnormality [47]

It is noteworthy that the Aβ sequence of WT rodents has a three amino acid difference compared to humans, making it less likely to aggregate and deposit into amyloid plaques [25]. Therefore, to study Aβ aggregation and plaque formation in rodents it is necessary to manipulate them genetically [25]. Most transgenic mouse models focus on overexpressing human APP, PS1, tau, or APOE variants.

3. Chronic Stress

3.1. Human Studies of Chronic Stress

Chronic stress has been implicated in the etiology of AD. The likelihood of developing AD has been shown to increase by a factor 2.7 with the personality trait distress proneness [25, 48, 49]. Moreover, AD patients show elevated plasma cortisol levels [50, 51] with higher levels of plasma cortisol being associated with a more rapid disease progression and cognitive deterioration [51, 52].

Sustained elevated levels of glucocorticoids can cause volumetric and dendritic changes in the hippocampus of rats, mice, and tree shrews [5356], decrease neurogenesis, and impair long-term potentiation [53, 57, 58]. It has, therefore, been proposed that alterations in HPA-axis functioning might also contribute to the etiology of AD [5961].

Evidence from studies over the last 20 years indicates that major depression may serve as a risk factor for developing AD [6269]. A lifetime history of depressive episodes doubles the chance of developing AD [70]. Interestingly, patients with major depression show a cerebrospinal fluid (CSF) profile of Aβ-species that resembles the profile seen in AD. They display decreased levels of Aβ42 and a decreased Aβ40 : Aβ42 ratio [71], which are considered putative biomarkers for AD [72]. In addition, the severity of depression correlated with binding of 2-(1-{6-[(2-{18F}Fluoroethyl)(methyl)amino]-2-naphthyl}ethylidine)malononitrile, also known as FDDNP, a tracer that binds to plaques and tangles, in the temporal lobe [73]. Moreover, more plaques and tangles in the hippocampus as well as a more rapid cognitive decline have been observed in AD patients with a lifetime history of major depression compared to patients without such history [74]. In contrast, others have suggested that major depression does not function as an independent risk factor for AD, but should merely be viewed as an AD prodrome [63, 75, 76].

3.2. Animal Studies of Chronic Stress

Several paradigms have been used to model the effects of chronic stress in mouse models of AD. The paradigms that have been applied most frequently are chronic isolation stress and chronic restraint or immobilization stress. Table 2 summarizes the current evidence for effects of stress exposure in transgenic mouse models of AD.

Table 2.

Effects of stress exposure in transgenic mouse models of Alzheimer's disease.

Mouse model Exposure Duration of the experiment Age at the start Effects on the brain Effects on behavior Reference
Tg2576 Chronic isolation stress 3 months From weaning ↑ soluble Aβ40 (38%) and Aβ42 (59%) in hippocampus, no change in Aβ40 : Aβ42 ratio, no difference in APP, α- or β-CTF levels, no changes in IDE, NEP (neprilepsyn) or APOE levels Not measured [79]

Tg2576 Chronic isolation stress 5 months From weaning ↑ Aβ plaques
↓ proliferation in DG
↓ contextual memory at 6 months [77]

Tg2576 Chronic isolation stress 6 months From weaning ↑ Aβ40 + Aβ42 levels and plaque deposition in neocortex and hippocampus
↑ expression of GR and CRFR1 in neocortex and hippocampus
↑ basal corticosterone in plasma
Not measured [78]

TASTPM Repeated novel cage exposure (1 h/day, 4x/week) 5 weeks 4 months No changes in basal corticosterone levels
↓ soluble Aβ40 levels in the frontal cortex + hippocampus
↓ insoluble Aβ42 levels in frontal cortex + hippocampus, no difference in endocannabinoid levels in frontal cortex and hippocampus
No difference in locomotion, nor in anxiety levels contextual memory ↑ [80]

APPswe Acute restraint stress (for 4 h) 4 hours 19 months ↑ 175% in blood glucose levels, dropping to below basal values 2 hours after restraint
↑ in stress-induced corticosterone release
Not measured [81]

Tg2576 Acute restraint stress (for 3 h) 3 hours 3-4 months ↑ interstitial fluid Aβ, no difference in APP or β-CTF levels
α-CTF levels, no changes in IDE, NEP (neprilysin) or APOE levels
Not measured [79]

APPV717I-CT100 Chronic immobilization stress (6 h/day, 4x/week) 8 months 3 months ↑ Aβ plaques in hippocampus, entorhinal + piriform cortex
↑ APP-CTFs
↑ pyknotic cells in hippocampus + entorhinal cortex
↑ phospho-tau in CA3 + entorhinal cortex
↑ Corticosterone in plasma
↑ cognitive impairment [82]

Tg2576 Chronic immobilization stress (6 h/day, 4x/week) 6 months 3 months Not measured ↓ cued food preference [82]

Tg2576 Chronic restraint stress (2 h/day) 16 days 14 months ↑ Aβ plaques in hippocampus, PFC, cingulate, motor, parietal and piriform cortex
↑ Aβ40 + Aβ42 in cortical homogenates
↑ immunoreactive astrocytes near plaques
↑ phospho-tau
↓ dendritic arborization of cortical neurons
↓ MMP-2 (Aβ-degrading enzyme)
↑ basal corticosterone levels
Not measured [83]
PS1-L286V Chronic restraint stress (6 h/day) 3 or 15 weeks 7 weeks 3 weeks exposure
↓ body weight
↑ adrenal gland weight
↑ corticosterone levels in plasma
↑ number of degenerating neurons in DG, CA3, and retrosplenial cortex, no effect on number of granule neuron precursors (Pax6) or proliferating cells (Ki67) in DG and/or SGZ
↓ BrdU-positive cells
↑ DCX-positive neuronal progenitor cells
15 weeks exposure
↓ body weight
↑ adrenal gland weight
↑ number of degenerating neurons in DG, CA3 and retrosplenial cortex, no effect on # of granule neuron precursors or proliferating cells in DG and/or SGZ
Not measured [84]

3xTg Dexamethasone administration (1 or 5 mg/kg) i.p. 7 days 4 months ↑ Aβ in hippocampus, neocortex, amygdala
↑ tau in dendrites and axons in hippocampus, neocortex, amygdala
↑ insoluble Aβ40 and Aβ42
↑ total APP, BACE1, C99 levels
↑ basal corticosterone levels from 9 months on
Not measured [85]

Chronic isolation stress by subjecting mice to either 3, 5, or 6 months of social isolation from weaning, has thus far only been used in the Tg2576 mouse model of AD [7779]. This resulted in elevated levels of soluble Aβ40 and Aβ42 up to 59% and increased plaque deposition in the hippocampus and the neocortex [77, 78]. Moreover, this stress exposure paradigm caused a rise in basal plasma corticosterone levels, paralleled with an increased expression of the glucocorticoid receptor (GR) and corticotropin-releasing factor (CRF) receptor 1 [78]. In addition, impaired contextual memory and decreased cell proliferation in the hippocampal dentate gyrus was observed. Interestingly, the effects of isolation stress on memory deficits and cell proliferation in the dentate gyrus could be prevented by a 14-day treatment of fluoxetine [77].

Another widely used stress paradigm is restraint stress. Acute short-term restraint stress elevated intracerebral interstitial Aβ levels in Tg2576 mice [79] and stress-induced corticosterone release in APPswe mice [81]. Administering CRF or a CRF-antagonist indicated that the interstitial rise in Aβ depended on CRF levels [79]. Acute restraint stress furthermore resulted in a 175% increase in blood glucose levels in APPswe mice, suggesting a wide impact on metabolism [81].

Chronic restraint stress has so far been performed in 3 different mouse models of AD: APPV717I-C100, Tg2576, and PS1-L286V mice. Applying chronic restraint stress to APPV717I-C100 and Tg2576 mice generally resulted in an increased Aβ plaque load, increased Aβ40 and Aβ42 levels, increased tau phosphorylation and increased basal plasma corticosterone levels [8284]. Chronic restraint stress applied to APPV717I-CT100 mice additionally induced cognitive impairment as measured for example by using cued food, that is, powdered chow mixed with a certain aroma, in the social transfer of food preference task [82]. Chronic restraint stress has also been associated with neuropathological alterations in AD mouse models. PS1-L286V mice exposed to chronic restraint stress displayed elevated numbers of degenerating neurons and a decreased number of proliferating cells in the hippocampus as compared to nonexposed mice [84]. Chronic restraint stress in APPV717I-CT100 mice caused elevated numbers of pyknotic cells in the hippocampus [82] and reduced dendritic arborization of cortical neurons in Tg2576 mice [83].

Another method to assess the effects of stress is by mimicking the physiological stress response by administering synthetic glucocorticoids, such as dexamethasone, for 7 days. Application of this approach in 3xTg mice resulted in elevated Aβ- and tau-immunoreactivity in the hippocampus, amygdala and neocortex and increased levels of insoluble Aβ40 and Aβ42 and total APP, β-site of APP cleaving enzyme (BACE1) and APP fragment C99 levels in brain homogenates [85].

4. Environmental Enrichment

4.1. Human Studies of Environmental Enrichment

A reduced risk for developing and a slower rate of cognitive decline have been observed in people having a greater purpose in life and higher levels of physical activity [86, 87].

4.2. Animal Studies of Environmental Enrichment

In the field of animal research, the environmental enrichment (EE) paradigm is frequently used to manipulate physical activity and social interactions. By introducing mates (social interaction) and/or toys (physical activity) into the cage of the rodent [88], this paradigm stimulates cognition as well as sensory and motor behavior with concomitant intracerebral cellular and molecular changes [89, 90]. To examine the effect of EE in AD several different paradigms have been imposed on various mouse models of AD. Table 3 summarizes the effects of EE in transgenic mouse models of AD.

Table 3.

Environmental enrichment in transgenic transgenic mouse models of Alzheimer's disease.

Mouse model Exposure Duration of the exposure Age at the start Effect on brain Effect on behavior Reference
APPswe Enriched housing (multiple mice in a large bin containing an inner cage with platforms, passageways, running wheels, toys, and novel habitats) + novel complex environment 3x weekly for several hours 4 months 16 months No differences in total Aβ load
Improved MWM performance [91]

TgCRND8 Enriched housing (equipped with diverse physically and cognitively stimulating objects, for example, gnawing wood, tunnels, balls, running wheels, and ladders) 4 months 1 month ↑ angiogenesis
↑ ApoE, LRP1, A2M
↓ RAGE
Not measured [92]

TgCRND8 Enriched housing (equipped with diverse physically and cognitively stimulating objects, for example, gnawing wood, tunnels, balls, running wheels, and ladders) 4 months 1 month ↑ BrdU-positive cells
↑ synaptophysin immunoreactivity in the hippocampus
Not measured [93]

TgCRND8 Enriched housing (plastic inset, wooden climbing frame, and a nesting material) 4 months 1 month Not measured ↑ exploratory behavior
↓ anxiety-related behavior
No effects in learning and memory,
as assessed with MWM, barrier test, open-field, elevated plus maze, object recognition task, and Barnes maze
[94]

APP23 Enriched environment (multiple mice housed in large cages with a rearrangeable system of plastic tubes and cardboard boxes) 1 months 6, 18 months No differences in plaque load
↑ DCX/CR ratio
↑ DCX- and calretinin-positive neurons in the hippocampus
[95]

APP23 Enriched housing (spacious cage equipped with a rearrangeable system of tubes, a cardboard box house, wire mesh ladders, and a crawling ball) 9 months 2 months No differences in plaque load in neocortex or hippocampus
↑ hippocampal neurogenesis (DCX, calretinin)
↑ BDNF, NT-3 in the hippocampus
Improved learning and memory in MWM [96]

APPswe/PS1ΔE9 Enriched housing (multiple mice in a large cage with crawl-tubes, platforms, running wheels and toys, changed weekly) 6 months 1.5 month ↓ total Aβ deposition, 28% in the hippocampus and 36% in entorhinal cortex
↑ synaptophysin in CA1 and CA3
Improved performance in MWM, RAWM [97]
APPswe/PS1ΔE9 Enriched environment (large cages, running wheels, colored tunnels, toys, and chewable material) 1 month, 3 hours daily, next 4 months three times per week 1 month ↓ neocortical and hippocampal Aβ deposits
↑ increased neprilysin expression
Not measured [98]

PS1/PDAPP Enriched housing (multiple mice in a large bin containing an inner cage with platforms, passageways, running wheels, toys, and novel habitats) + novel complex environment 3/weekly for several hours 5 months weaning ↑ gene expression of TTR, NF-κB inhibitors, Improved performance in MWM, RAWM and platform recognition tasks [99]

APPswe/PS1ΔE9 Enriched housing (larger cages with running wheels, plastic play tubes, cardboard boxes, and nesting material that were changes or rearranged weekly) 6 months 2 months ↑ 68% of plaque area in the hippocampus
↑ 52% of total Aβ in hippocampus
Not measured [100]

APPswe/PS1ΔE9 Enriched housing (larger cages with running wheels, plastic play tubes, cardboard boxes, and nesting material that were changes or rearranged weekly) 6 months 2 months ↑ 50% Aβ42 in the hippocampus
↑ 25% in hippocampal plaque load
Improved performance in MWM, RAWM [101]

APOE3, APOE4 Cages with exploratory objects (toys, tunnels, and running wheels) 5 months 3 weeks improvement in T-maze performance in APOE3 only ↑ expression of NGF
↑ Synaptophysin in the hippocampus of APOE3 only
[102]

APOE3, APOE4 Enriched housing ( cage with running wheel, labyrinth, bedding, house, chains, and wooden blocks) 5 months 3 weeks ↑ hippocampal Aβ deposits in the APOE4 Not measured [103]

APOE3, APOE4 Wheel running 6 weeks 10 months ↑ BDNF in both
↑ TrkB, PAK and synaptophysin only in APOE4
Improved performance in place recognition in both genotypes
Improved RAWM in APOE4 only
[104]

4.2.1. APP Mice

EE, in terms of housing multiple mice in a larger cage with platforms, running wheels, toys, and other novel habitats, for a period of 6 months improved cognition in a battery of tests such as Morris water maze (MWM), circular platform, platform recognition and radial arm water maze, despite signs of stable Aβ deposition in 16-month-old APPswe mice [91]. EE for 4 months in 5-month-old TgCRND8 mice did not significantly alter soluble levels of Aβ in the brain or the blood, but did enhance mRNA expression of angiogenic genes [92]. EE in this mouse model attenuated age-related reductions in cell proliferation, neurogenesis and synaptic plasticity [93], while the same paradigm in another laboratory elevated Aβ plaque load without compromising behavioral phenotypes such as feeding and drinking pattern, grooming, locomotion or cognition [94, 105]. In an attempt to disentangle the exact components of EE that influence phenotypes in APP mutant mice, Wolf et al. [96] exposed APP23 transgenic mice to either an enriched environment or unlimited access to a running wheel and compared both conditions with standard housing. EE had differential effects upon improving performance in the MWM as compared to the increased physical activity and standard housing groups, however, no differential effects on plaque load in the neocortex or hippocampus were found [95, 96]. Moreover, mice exposed to EE exhibited signs of increased hippocampal neurogenesis and neurotrophic support [95, 96].

4.2.2. APP/PS1 Mice

When comparing social interaction and physical activity, differential effects of EE can be observed on learning and memory processes, Aβ plaque load and synaptophysin immunoreactivity of 9-month-old APP/PS1 transgenic mice [97]. EE in APPswe/PS1ΔE9 mice reduced cortical and hippocampal Aβ deposition with mice being more active in the running wheel showing an even more marked decrease in Aβ [98]. Furthermore, EE in PS1/PDAPP mice attenuated cognitive impairments [99].

Possibly in contrast with overt beneficial effects, 2-month-old female APPswe/PS1ΔE9 mice exposed to EE for several months displayed increased Aβ levels in the neocortex, and hippocampus [100]. After further backcrossing these mice to a C57Bl6 background strain in order to attain fewer genetic background differences, the same group demonstrated that EE in 2-month-old transgenic APPswe/PS1ΔE9 female mice, attenuated cognitive deficits [101], but still exhibited a 25% increase in Aβ deposits in cortical and hippocampal brain regions [101]. One could argue that enhanced secretion and deposition of toxic soluble Aβ species (scavenging the toxic species into packages away from intracellular and synaptic compartments) may be a mechanistic explanation for these findings.

4.2.3. APOE Mice

EE in mice carrying the APOE3 allele improved learning and memory, as assessed with the T-maze test, while it had no effect in the ones carrying the E4 allele. The improved cognitive performance in APOE3 mice was associated with increased neocortical and hippocampal synaptophysin- and nerve growth factor-immunoreactivity, which was not observed in the APOE4 mice [102].

In conclusion, the majority of studies indicate that EE affects AD-related phenotypes in transgenic mouse models of AD pathology, mostly in a beneficial manner, particularly with regards to behavior. However, contradictory results have been reported which can possibly be explained by different experimental paradigms, age, sex, and genetic background of the mice used.

5. Metal Exposure

5.1. Lead

Lead exposure has been proposed as a risk factor for AD by some authors [106, 107] while others have argued against it [108]. No studies to date have performed lead exposure experiments in mouse models of AD although other animal work has indicated that lead exposure early in life may contribute to the onset of AD-related pathology later in life [109, 110].

5.2. Aluminum

While it has been proposed that occupational aluminum exposure is not a significant risk factor for AD [108], prolonged exposure to aluminum in drinking water is significantly associated with an increased risk of developing AD in a dose-dependent manner with the relative risks varying from 1.00 to 2.14 (for review see [111]). These findings should be regarded with some caution as aluminum concentrations varied highly between the different studies and many variables (such as interaction with other chemical constituents in the drinking water as well as alternative sources of aluminum, for example through antacid use or dietary intake) have often been overlooked in these studies [111].

Products made of baking-powder often contain high levels of aluminum, and it has been observed that AD patients were more frequently exposed to ingestion of foods containing baking-powder (retrospectively investigated) than age-matched controls [112]. Other studies suggested that AD patients have significantly enhanced gastrointestinal absorption of aluminum (up to 1.64 times higher) compared to age-matched controls, and indicate that differential gastrointestinal function may lead to a systemic rise of aluminum [113, 114].

Praticò and colleagues [115] reported that Tg2576 mice exposed chronically to dietary aluminum displayed increased Aβ40 and Aβ42 levels, plaque deposition, and markers of oxidative stress in the hippocampus and neocortex compared to non-exposed Tg2576 mice. Others authors, however, were not able to replicate these findings. They reported that chronic aluminum treatment in Tg2576 mice did not affect Aβ load in the cerebral cortex or oxidative stress reactions in the hippocampus, nor impair spatial cognition, as measured by the MWM [115117]. Aluminum treatment did raise the levels of aluminum and other metals in the hippocampus, neocortex and cerebellum, but no major differential effects could be found between Tg2576 and WT mice [116, 118]. The differential effect of aluminum exposure in these mice could possibly be explained by higher concentrations of aluminum in the chow, differential ages at the start of the experiment and a shorter duration of exposure.

5.3. Iron, Zinc, and Copper

The endogenous biometals iron, zinc and copper have often been implicated in AD, as they are present in and around amyloid deposits in the AD brain and their presence can promote aggregation of Aβ [119121].

Although no report has confirmed a direct link between iron exposure and the risk of developing AD, Dwyer and colleagues do propose a ferrocentric model of AD [122]. Higher levels of ferritin iron in the basal ganglia have been considered a risk factor for AD [123, 124]. AD patients show elevated levels of iron in the hippocampus [125], and this metal seems to concentrate in the core and rims of plaques in the amygdala [126].

Rodent research has indicated that gestational or early developmental iron deficiency can alter the expression of the APP and CLU genes implicated in synaptic plasticity, dendritic outgrowth, and AD pathogenesis [127129]. Neonatal administration of iron for 3 days to APPswe/PS1ΔE9 mice was found not to alter Aβ deposition in the hippocampus and temporal cortex at 6 months of age but did cause changes in lipid composition, decreased steady-state levels of oxidative damage markers, and increased astrocyte levels in the temporal cortex [130].

Zinc seems to play a double role in AD etiology. Low levels of zinc have been reported to be protective against Aβ formation [131] and metalloproteases, such as neprilysin and insulin-degrading enzyme (IDE), that degrade Aβ are zinc dependent [132]. It has also been found that high levels of zinc elevate Aβ toxicity [131, 133] and promote total Aβ aggregation [121]. AD patients displaying higher levels of zinc in hippocampus and amygdala [125, 126] exhibited normal zinc serum levels, but significantly lower zinc levels in CSF compared to matched controls [134]. This may be explained by the binding of zinc to Aβ in the brain parenchyma [135]. However, to our knowledge, no reports have been published on putative associations between zinc exposure and risk of AD in the human population. Nonetheless, several studies have investigated the effects of altered zinc intake in AD mouse models.

Stoltenberg et al. [136] reported that lowering zinc by a 3-month dietary deficiency increased the plaque load in APPswe/PS1ΔE9 mice by 25%, without changing zinc ion distribution, zinc transporter mRNA expression levels nor inducing oxidative stress [136]. Alternatively, administrating zinc to TgCRND8 and Tg2576 mice through the drinking water for a period of 5 and 9 months, respectively, lowered the amyloid plaque burden in the hilar and molecular region of the dentate gyrus, while impairing spatial memory in MWM [137]. Concurrently, long-term administration of high zinc concentrations in TgC100 mice did not significantly affect soluble Aβ levels or levels of glial fibrillary acidic protein (GFAP), superoxidase dismutase 1, APP, β-secretase-cleaved carboxyl-terminal fragment, or neurofilament 200, a marker for neuronal damage [138]. Interestingly, a genetic reduction of zinc in the brain of Tg2576 mice, by crossing these mice with a zinc transporter 3 deficient mouse (ZnT3−/−), significantly reduced the plaque load in the hippocampus and neocortex while increasing the ratio between soluble versus insoluble Aβ [139].

AD patients have been shown to display reduced copper levels in the amygdala and hippocampus, while copper levels are specifically elevated in amyloid plaques [125, 126]. Copper intake in AD patients decreases the reduction of Aβ42 in CSF most typically seen as the disease progresses, but does not ameliorate cognitive performance [140]. Adding copper to drinking water of cholesterol-fed rabbits causes accumulation of Aβ and the formation of plaque-like structures [141].

Exposing AD mouse models to chronic upregulation of copper has yielded conflicting results. Chronic copper administration to APP715SL mice did not alter copper, zinc, iron, Aβ nor APP levels in the brain [142]. Long-term administration of high levels of copper resulted in a 18% decrease in soluble Aβ40 and increased zinc levels in the brain without changing GFAP, SOD1, APP, C100, or NF200 levels to TgC100 mice. Yet, copper exposure in 3xTg mice led to elevated steady-state levels of APP, and C99 as well as to increased Aβ production and tau phosphorylation in the brain [143]. Interestingly, copper, APP and Aβ seem to be closely connected; Tg2576 mice displayed an overall reduction of copper in the brain whereas the ablation of APP and amyloid precursor-like protein 2 increased overall central copper levels [144, 145].

In summary, both human and rodent research on the exact contributing roles of metal exposures in interaction with AD risk genes APP and PS1 remain largely inconclusive. For an overview of metal exposure in the transgenic animal models of AD listed above, see Table 4.

Table 4.

Environmental exposure to metals and electromagnetic fields in transgenic mouse models of Alzheimer's disease.

Mouse model Exposure Duration of the exposure Age at the start Effect on brain Effect on behavior Reference
TgV337M Aluminum-mltolate i.p. injection at various concentrations (50–100–200 μM) Max 14 days 3 months Al levels were too low to induce changes in tau phosphorylation in brain homogenates, but Al concentration was lethal Not measured [146]

Tg2576 Dietary aluminum (2 mg/kg diet) 9 months 3 months ↑ soluble and insoluble Aβ40 and Aβ42 in neocortical and hippocampal homogenates
↑ plaque load in hippocampus and neocortex
↑ oxidative stress markers
Not measured [115]

Tg2576 Dietary aluminum lactate (1 mg/g diet) 120 days 5 months No significant differences in Aβ40 and Aβ42 in cortical homogenates, no alterations in proliferation, survival or differentiation of BrdU-positive neurons in DG No improvement MWM [117]

Tg2576 Dietary aluminum lactate (1 mg/g diet) 6 months 5 months ↑ Al concentration in hippocampus and cerebellum
↑ Cu in hippocampus
↓ Fe in cerebellum
↑ Mn and Zn in neocortex, hippocampus and cerebellum
Not measured [118]

Tg2576 Dietary aluminum lactate (1 mg/g diet) 6 months 5 months ↑ Al concentrations in the hippocampus, but no difference between WT and Tg animals, no difference in oxidative stress reaction in the hippocampus between WT and Tg Not measured [116]

APPswe/PS1ΔE9 Iron carbonyl (1 mg/ml) 3 days P12 No difference in Aβ plaque load in hippocampus and temporal cortex, no difference in microglial activity
↑ GFAP levels in temporal cortex
↑ saturated fatty acids
↓ unsaturated fatty acids
↓ oxidative damage markers
Not measured [130]

APPswe/PS1ΔE9 Zinc-deficient (<10 parts Zn per million (ppm)) 3 months 9 months No significant difference in serum zinc levels, no difference cortical volume
↑ 25% in total plaque volume, no difference in number of plaques or laminar distribution, no difference in oxidative stress markers
Not measured [136]

Tg2576 Zinc in drinking water (10 ppm/0.153 mM Zn) ±12 months From conception ↓ Aβ deposits in hilar and molecular region of the DG ↓ spatial memory in MWM both in Tg and WT, buth most pronounced in Tg [137]

TgCRND8 Zinc in drinking water (10 ppm/0.153 mM Zn) 5 months From weaning No significant differences ↓ spatial memory in MWM both in Tg and WT, buth most pronounced in Tg [137]
TgC100 Zinc in diet (ZnSO4, 1000, 500 or 300 ppm) 15 months 7 weeks ↑ Brain Zn levels in brain homogenates
↓ Cu levels (n.s.)
↓ Cu/Zn ratio
↓ 13% soluble Aβ 40 (trend)
No changes in GFAP, SOD1, APP, C100, nor NF200 (neuron loss), no difference in intensity or distribution of Aβ or GFAP staining
Not measured [138]

TgC100 Copper in diet (CuSO4 150 or 100 ppm) 7 weeks 9 months No significant differences in Cu levels in brain homogenates
↑ Zn levels
↓ 18% soluble Aβ 40, no changes in GFAP, SOD1, APP, C100, nor NF200 (neuron loss), no difference in intensity or distribution of Aβ or GFAP staining
Not measured [138]

3xTg Copper sulfate (250 ppm) in 5% sucrose drinking water 3 or 9 months 2 months 3 months exposure:
↑ steady-state levels APP, C99 and BACE1
↑ Aβ 40 in total plaque load in hippocampus, no alterations in total tau, phospho-tau nor Thy1.2 transcription activity
↑ AT8-positive neurons in CA1, no changes in steady-state levels of cdk5, p35/p25, GSK-3β or phospho-GSK-3β
↓ SOD1 activity in brain homogenates
9 months exposure:
↑ steady-state levels APP, C99, C83, BACE1, ADAM10
↑ soluble Aβ40, phospho-tau, no alterations total tau levels
↑ p25 formation
↓ SOD1 activity
Not measured [143]

AβPPsw Electromagnetic field exposure (918 MHz, 0.25 W/kg ± 2 dB) 2 × 1 h p/d 7-8 month exposure 2 months
5 months
Young adult 7 months exposure, no significant differences in soluble Aβ in hippocampus + neocortex, no effect op hippocampal DNA repair enzymes, antioxidant enzyme markers, protein oxidative damage, nor striatal DNA oxidation
Aged adult 8 months exposure
↓ Aβ plaque load in hippocampus (−35%) and entorhinal cortex (−32%)
↑ soluble Aβ in hippocampus + neocortex
Young adult 7 months exposure
Prevention of cognitive deficits in retroactive interference
↑ Y-maze spontaneous alternation level
No differences in open field activity, balance beam, string agility, and elevated plus maze
Aged adult 8 months exposure
Reversal of cognitive deficits
[27]

6. Traumatic Brain Injury

6.1. Human Studies of Traumatic Brain Injury

Traumatic brain injury (TBI) has repeatedly been identified as a risk factor for AD. It has been suggested that TBI accelerates the onset of AD and that the severity of the injury increases the risk of AD [147]. AD-like pathology has been observed after acute brain trauma, even in brains of young individuals. A polymorphism in the promoter of the gene that encodes neprilysin, causing a greater length in GT repeats, has been associated with the acute development of plaques following TBI [148]. In addition, carriers of the APOE4 genotype have been associated with poorer outcome after TBI [147].

6.2. Animal Studies of Traumatic Brain Injury

For an overview of TBI in mouse models of AD, see Table 5. After corticol contusion, 10- to 16-month-old PDAPP mice did not show significant differences in behavior or Aβ neuropathology following TBI, as compared to WT controls that underwent the same procedure of experimental brain injury [149]. Inducing TBI in the PDAPP mouse model at 4 months of age, accelerated memory loss as assessed with the MWM test. TBI also resulted in hippocampal neuronal loss one week after injury, which was associated with an increase in hippocampal Aβ40 and Aβ42 [150]. Furthermore, TBI resulted in long-term effects at 2, 5, and 8 months after TBI: a significant reduction in Aβ plaque load was found which was accompanied with more pronounced hippocampal atrophy. TBI induction in APPNLh/NLh mice caused a twofold increase in soluble hippocampal Aβ levels at 3 and 7 days after TBI. Additionally, post-TBI administration of caspase-3 inhibitors and the hypolipemic simvastatin were able to attenuate impaired hippocampal synaptic function, microglial activation and MWM performance after TBI induction in APPNLh/NLh mice [151, 152]. TBI induced by cortical impact provoked gene expression changes in 22-month-old APPswe mice compared to WT mice. Expression changes were detected in genes involved in various biological pathways such as immune response, cell cycle and cell death, cellular development, tissue development and connective tissue function and development, cellular movement, and hematological systems [153].

Table 5.

Traumatic brain injury in transgenic mouse models of Alzheimer's disease.

Mouse model Exposure Duration exposure Age at start exposure Effects on brain Effects on behaviour Reference
APP-YAC Cortical contusion impact (3-mm diameter impounder onto the left parietal cortex, 100 ms; velocity (v) = 4.8−5.2 m/s; depth = 1 mm) 10–16 months No difference No difference in MWM [149]

PDAPP Cortical impact brain injury (3-mm diameter impounder onto the left parietal cortex, 100 ms; v = 4.8−5.2 m/s; depth = 1 mm) 4 months Increased hippocampal neuronal death MWM memory impairment in transgenic as compared to controls [150]

PDAPP Controlled cortical impact (3-mm diameter impounder onto the left parietal cortex, 100 ms; v = 4.8−5.2 m/s; depth = 1 mm) 6 months Hippocampal atrophyDecrease the hippocampus and cingulate cortex 3 months after TBI Not measured [156]

PDAPP Controlled cortical impact (3-mm diameter impounder onto the left parietal cortex, 100 ms; v = 4.8−5.2 m/s; depth = 1 mm) 24 months Increased hippocampal neuronal loss and gliosis Regression of Aβ in the hippocampus Not measured [157]

APPswe Controlled cortical impact (3-mm diameter impounder onto the left parietal cortex, 47 ms; v = 5.82 m/s; depth = 1.2 mm, driving pressure 73 psi) 3 months 2× increase in Aβ40 and Aβ 42
Reduced CA3 ynaptophysin immunoreactivity
MWM performance deficit [152]

Tg2576 Controlled cortical impact (mild to moderate, 2-mm diameter impounder onto the right cortex, v = 3.3 m/s; depth = 1 mm) 22 months Gene expression differences in inflammation, immune response and cell death Not measured [153]

Tg2576 Controlled cortical impact (3-mm diameter impounder onto the left parietal cortex, 100 ms; v = 4.8−5.2 m/s; depth = 1 mm) Repetitive (2×) 9 months Increased hippocampal amyloid deposition MWM cognitive dysfunction [154]

Single and repetitive mild TBI, using a cortical impact device, in 9-month-old Tg2576 mice as compared to sham treated transgenic and WT, resulted in significant cognitive dysfunction (measured with MWM) without affecting motor performance 16 weeks after TBI. However, only repetitive TBI caused increased Aβ burden in the hippocampus and neocortex with a parallel increase in isoprostane, an indicator for increased oxidative stress [154]. TBI in 10-month-old transgenic mice overexpressing either human APOE3 or 4, was associated with differential gene expression, particularly in genes related to oxidative stress, with an increased expression of antioxidant genes in the APOE3 mice as compared to the APOE4 [155].

Thus, accumulating evidence indicates that TBI interacts with AD-related genes.

7. Electromagnetic Field Exposure

Occupational exposure to electromagnetic field (EMF) has been proposed as a risk factor of AD. In particular, extremely low-frequency exposure has been implicated to increase the odds to develop AD up to 2.03 (as reviewed in [155]). Strikingly, Arendash et al. [27] demonstrated that long-term high-frequency exposure to EMF (i.e., similar to that generated by cell-phone use) was beneficial to AβPPswe mice (see Table 4). EMF exposure in young adult AβPPsw mice prevented the age-related genotype-specific cognitive impairment, while EMF in aged AβPPsw mice was also able to reverse cognitive impairment in these animals. Chronic EMF exposure furthermore influenced Aβ aggregation in the brain, with higher levels of soluble Aβ and less Aβ plaques in the hippocampus and entorhinal cortex of AβPPsw mice. EMF exposure has been proposed to contribute to a decrease in Aβ aggregation, via altering levels of transthyretin [158]. Transthyretin is known to sequester Aβ in CSF, thereby hindering its aggregation into amyloid plaques [158]. Interestingly, AD patients show a significant decrease in CSF transthyretin levels [159] while decreased transthyretin levels have also been found in blood serum of long-term wireless phone users [158]. Thus, effects of EMF are quite puzzling while the association with AD remains to be firmly established.

8. Effects of Diet and Nutritional Factors

8.1. Mediterranean Diet

Various dietary and nutritional factors seem to be protective or detrimental in the development and course of AD. One of the most prominent is the Mediterranean type of diet which has been linked to reduced risk of developing AD and showing a dose-response effect (high adherence to Mediterranean diet, OR: 0.76; moderate adherence, OR: 0.47) [160162].

A typical Mediterranean diet is characterized by higher consumption of vegetables, fruits, cereals, fish, and olive oil and associated with a general higher consumption of unsaturated fatty acids and lower consumption of saturated fatty acids, usually accompanied by mild or moderate alcohol intake (preferably red wine taken with meals) [160]. The exact factors and mechanisms by which the Mediterranean diet is protective remains to be elucidated, although it has been speculated that this diet can attenuate the detrimental effects of oxidative stress and inflammation [161].

8.2. Western Diet and Obesity

Obesity during mid-life is associated with an increased risk for AD, with an OR of 2.4, additively increasing up to 6.2 when combined with high total cholesterol levels and high systolic blood pressure [163]. Higher intake of calories and fat have been associated with increased risk for developing AD, particularly in APOE4 carriers, with a hazard ratio of 2.3 [164]. Western, high-fat and low carbohydrate diet for 4 months in 1-month-old Tg2576 mice, increased levels of soluble Aβ in brain homogenates, while the treatment did not have any effect on plaque load [165]. Further, insulin resistance induced by 5 months of high-fat diet, in 9-month-old Tg2576 mice, was associated with to a twofold increase of Aβ40 and Aβ42 peptide content in the hippocampus and a twofold increase in plaque burden in the neocortex, with a concomitant acceleration of cognitive decline as measured by the MWM. In addition, γ-secretase activity was increased, while the expression of IDE was decreased by this diet [166]. APP/PS1 KI mice exposed to Western, high-fat diet showed increased oxidative stress markers as measured in brain homogenates of 2-month-old mice when compared to nontransgenic controls, but Aβ levels were not altered [167]. In another study, Western diet increased Aβ deposition in the hippocampus of the APPswe/PS1ΔE9 transgenic mice at 18 months of age, after a period of 12 months on a high-fat diet [168]. In the 3xTg mouse model of AD, a high fat diet starting at the age of 4 months for a total period of 13 months, induced similar effects in the frontal cortex [169].

8.3. Cholesterol

As the generation, deposition, and clearance of Aβ is regulated by cholesterol, many studies have specifically focused on the implication of lipids, cholesterol metabolism, related vascular disease, APOE genotype, and their interrelationships on the development of AD [170172]. The precise mechanisms underlying cholesterol and APOE4 need further investigation, as it is not clear whether cholesterol and the APOE4 genotype act as independent factors or interact with one another or whether the effect of APOE4 is partially mediated by high cholesterol levels [171174]. Also, hypercholesterolemia in 3-month-old APPswe/PS1M146L mice has been shown to accelerate Aβ accumulation while drug-induced hypocholesterolemia reduced the amyloid pathology [175, 176].

8.4. Docosahexaenoic (DHA)

Studies in mouse models of AD amyloidosis, such as Tg2576, APPswe/PS1ΔE9, and 3xTg, have shown that a diet rich in the omega-3 fatty acid DHA reduces Aβ accumulation and somatodendritic tau accumulation, improves cognition, and induces cerebral hemodynamic changes [168, 177180]. Such findings are in line with evidence from epidemiological studies showing a protective effect of diets rich in omega-3 fatty acids [181183]. More specifically, DHA-enriched diet was shown to increase relative cerebral blood volume with a concomitant improvement in spatial memory and reduction of Aβ load in APPswe/  PS1M146L mice [184]. Exposing APPswe/PS1ΔE9 mice to a diet high in omega-3 fatty acids, however, neither improved cognition in APPswe/PS1ΔE9 mice nor reduced hippocampal Aβ, but increased omega-3 fatty acid levels in their brain [185]. Interestingly, high levels of omega-6 were linked to cognitive impairment [185].

8.5. Vitamins

Dietary deficiency of B6, B12, and folate for 7 months increased Aβ levels in the brains of 15-month-old Tg2576 mice, without altering APP, BACE-1, A disintegrin and metallopeptidase 10 (ADAM-10), nicastrin, IDE, APOE, or neprilysin [186]. Additionally, the same pattern of dietary vitamin B deficiency led to increased expression of PS1 via DNA demethylation of the promoter region of the encoding gene in brain homogenates of TgCRND8 mice [187]. In the brains of mice of the same animal model, vitamin B deficiency increased the levels of glycogen synthase kinase 3β (GSK3β) and reduced the activity of protein phosphatase 2A, which are both involved in the hyperphosphorylation of tau [188]. Furthermore, folic acid deficiency for 3 months in APPswe mice did not affect the Aβ plaque load, but induced neuron loss in the CA3 region of the hippocampus and enhanced hippocampal DNA damage, as compared to controls [189]. Besides B6 and B12, deficiency of B1, also called thiamine, exacerbated Aβ pathology via an upregulation of BACE1 in brains of Tg19959 mice [190].

Furthermore, dietary supplementation with the coenzyme Q10 for 2 months delayed hippocampal atrophy in 22-month-old APPswe/PS1Leu235Pro mice as compared to vehicle treated controls [36, 37], with concurrent reduction in plaque load [36, 37].

Deficiency of vitamin A has been implicated in Aβ accumulation, loss of long-term potentiation and memory impairment, while administration of its active metabolite retinoic acid for a duration of 2 months was able to rescue these deficits in the frontal cortex and hippocampus of 7-month-old APPswe/PS1ΔE9 transgenic mice [191].

8.6. Caffeine and Green Tea

Besides the various nutritional factors, other lifestyle habits have also been associated with AD. Longitudinal studies have shown that coffee and tea drinking are associated with decreased risk for cognitive decline, dementia and AD in various population samples [192, 193]. Another study showed a protective effect of caffeine only in women, with a relative risk of 0.49 [194], but a meta-analysis estimated an overall protective effect against dementia, with a relative risk of 0.84, though pointing out the large heterogeneity in the methods of the various epidemiological studies [195].

Acute and long-term caffeine consumption was recently shown to delay cognitive decline and lower Aβ pathology in the hippocampus of 15-month-old APPswe and APPswe/  PS1M146L mice, by suppressing β- and γ-secretase levels [196, 197]. Furthermore, oral or intraperitoneal administration of epigallocatechin-3 gallate, which is derived from green tea, for 2 or 6 months, exerted beneficial effects in APPswe transgenic mice, at the age of 14 months. The beneficial effects consisted of a reduction in Aβ pathology in the neocortex and hippocampus, with a parallel improvement of working memory [198200]. Furthermore, administration of the citrus-derived flavonoid luteolin and its analogue diosmin for a total of 30 days, significantly reduced Aβ pathology in the hippocampus and neocortex of 9-month-old Tg2576 mice. This effect was mediated via an inhibition of GSK3β, which increased PS1 phosphorylation [201].

8.7. Wine

Moderate red wine consumption has been shown to be beneficial. Cabernet sauvignon administration for 7 months in 4-month-old Tg2576 mice attenuated the cognitive impairment that is observed in these mice, in terms of spatial memory, when compared to ethanol-consuming and tap water Tg2576 controls. Cabernet sauvignon consumption decreased cortical and hippocampal Aβ plaque load in these mice, by promoting nonamyloidogenic processing in the direction of α-secretase cleavage [202]. Further, in vitro studies in hippocampal neuron cultures derived from Tg2576 mice, showed that the polyphenol extracts from the Cabernet sauvignon grapes increased the levels of α-secretase, which promotes the nonamyloidogenic cleavage of APP that reduced the levels of Aβ peptides [202]. Furthermore, consumption of the muscadine wine was proven to attenuate Aβ pathology in brains of 14-month-old Tg2576 mice, following a 10-month wine treatment, with a different mechanism of action. In this case, muscadine consumption reduced the aggregation of Aβ, with a parallel improvement in spatial memory [203]. The differential effect of the two types of wine was attributed to their distinct composition in polyphenolic compounds, which have a differential effect on APP processing [203].

8.8. Nicotine

Smoking in humans has been linked with increased risk for AD [204206], while nicotine administration for 6 months to transgenic mice carrying the APPswe mutation reduced the levels of insoluble Aβ in various brain regions of 15-month-old mice [207, 208]. Nevertheless, nicotine administration for 5 months exacerbated hippocampal tau pathology in 6-month-old 3xTg mice, increasing tau hyperphosphorylation and aggregation, in combination with an earlier onset of these tau-related changes compared to controls [209].

8.9. Caloric Restriction

Caloric restriction in animals has been found to prolong mean and maximum life span, reduce body fat, attenuate age-related molecular changes, and slow the decline associated with aging in various species [210]. In nonhuman primates, for example, caloric restriction prolongs lifespan and delays the onset of diseases, such as cardiovascular diseases, diabetes, and brain atrophy [211]. Restricting caloric intake by 40% for 6 weeks in APPswe and 14 weeks in APPswe/  PS1M146L mice has been shown to reduce the number and size of amyloid plaques by 40% and 55%, respectively, while also reducing the plaque-related astrocyte activation. These effects were observed in the neocortex and hippocampus [210]. Similar effects have been observed in the hippocampus of the 3xTg model after 14 months of caloric restriction [212].

8.10. Others

In line with studies showing a beneficial effect of nonsteroidal antiinflammatory agents on AD pathology in transgenic models of AD [213], the phytogenic curcumin (a major dietary component in India) administered for 6 months reduced levels of soluble and insoluble Aβ, plaque load, oxidative stress and inflammatory response in various brain regions of 16-month-old transgenic mice carrying the APPswe mutation [214]. Additionally, blueberry supplementation and Gingko biloba extract treatment have been found to improve memory deficits in APP/PS1 and Tg2576 mice without affecting amyloid plaque load [215, 216]. An overview of all nutritional and dietary factors influencing animal models of AD is given in Table 6.

Table 6.

Dietary, nutritional, and lifestyle habits and mouse models of Alzheimer's disease.

Mouse model Exposure Duration of the exposure Age at the start Effect on brain Effect on behavior Reference
APPswe/ind Caloric restriction, 40% 2 weeks 3 months ↓ 40% in cortical and hippocampal plaque load Not measured [210]

APPswe/PS1M146L Caloric restriction, 40% 4 months 2 months ↓ 55% in cortical plaque load Not measured [210]

3xTg Caloric restriction, 40% 7 or 14 months 3 months ↓ hippocampal Aβ40, 42 Improved open field activity
Improved performance in MWM
[212]

APP/PS1 KI Western (40% fat) diet 1 month 1 month ↑ oxidative stress markers (protein nitrosylation, protein carbonyls, and lipid hydroperoxides) Not measured [167]

APPswe/PS1ΔE9 DHA 12 months 6 months ↓ Aβ deposition in the vasculature of the cingulate gyrus Not measured [168]

3xTg Western (35% fat, low n-3 : n-6 PUFA ratio) 9 months 4 months ↑ cortical Aβ40, 42
↑ cortical tau
↑ cortical GFAP
Not measured [217]

Tg2576 High-cholesterol diet (5% cholesterol, 10% fat, 2% sodium cholate, and 5.2 kcal/g) 2 months 1 month ↑ soluble Aβ40, 42
↑ plaque number and size in neocortex and hippocampus
β-CTFs
Not measured [175]

3xTg DHA-rich diet (1.3 g/100 g) 3, 6, 9 months 3 months ↓ soluble Aβ40, 42
↓ PS1 levels
↓ somatodendric accumulation of tau
↓ tau phosphorylation (whole-brain homogenates)
Not measured [179]

Tg2576 DHA-rich diet (0.6% DHA) 3 months 19 months ↓ 38% insoluble Aβ in cortex
↓ Aβ42 49%
↓ Aβ40 47.5%
↓ plaques by 39% in hippocampus, 49% in parietal cortex and 47% in perirhinal cortex
Not measured [177]

APPswe/PS1ΔE9 Fish oil-based diet (0.4% DHA, 0.4% EPA, and 0.2% arachidonic acid) 4 months 6 months ↓ Aβ40, 42 levels in the hippocampus Not measured [178]

APPswe/PS1ΔE9 DHA rich diet (0.6% with safflower oil) 3 months 3 months ↓ plaque load by 27–30% in neocortex, ventral hippocampus, striatum, only in females
↑ hippocampal synaptotagmin and drebrin in females
Not measured [180]

APPswe/PS1ΔE9 DHA rich diet (0.4%, low-saturated fatty acids, high PUFA, and low n-6/n-3 ratio) 6–13 months 2 months ↓ rCBV in 8 months and
↑ in 15 months in cerebral cortex
No diet effects on open field, MWM, reverse MWM, 12 circular hole board tasks [184]
Tg2576 Folate, B6, and B12 deficiency 7 months 8 months ↑ Aβ40, 42 peptides in neocortex and hippocampus
↑ plaques in neocortex and hippocampus
Not measured [186]

TgCRND8 Folate, B6, and B12 deficieny 3 months 1 month ↑ PP2A, GSK3b mRNA
↑ tau phosphorylation
Not measured [188]

APPswe Folic acid deficiency 3 months 7 months 20% loss of neurons in CA3
↑ DNA damage in hippocampus, measured by DNA strand breaks
Not measured [189]

Tg19959 Thiamine deficiency 10 days 2 months ↓ KGDHC (α-ketoglutarate dehydrogenase complex)
↑ neuronal loss in thalamus
↑ TNF-a and antimalondialdehyde, markers for oxidative stressAcceleration of amyloid plaques,
↑ total and insoluble Aβ
↑ BACE1 levels
Not measured [190]

APPswe/PS1Leu235Pro CoQ10 supplementation (2400 mg/kg/day, oral) 2 months 19 months ↓ hemisphere and hippocampal atrophy Not measured [36]

APPswe/PS1ΔE9 Retinoic acid (20 mg/kg, i.p.) 2 months 5 months ↓ Aβ plaque levels in frontal cortex and hippocampus
↓ APP-CTFs, 70% neocortex, 50% hippocampus,
↓ tau phosphorylation
↓ GFAP
Improved MWM performance [191]

APPswe/PS1M146L Caffeine (0.3 mg/ml in H2O) 1 month 19 months ↓ of total Aβ, 46% in entrorhinal cortex, 40% in the hippocampus
↓ soluble Aβ42, neocortex 51%, hippocampus 59%
↓ soluble Aβ40, neocortex 25%, hippocampus 37%
↓cRaf-1 in hippocampus
Improved working memory in RAWM [196, 197, 218]

APPswe/PS1M146L Caffeine (0.3 mg/ml in H2O) 4 months 4 months ↓ insoluble Aβ42 32%, soluble Aβ40 37% in the hippocampus
↓ expression of PS1 and BACE1
Improved performance in MWM, RAWM, platform recognition [218]

APPswe/PS1M146L Caffeine (1.5 mg/0.2 ml, oral) 2 months 15–20 months ↓ soluble Aβ 42 in neocortex and hippocampus, by 51% and 59%, respectively Improved RAWM performance [197]

Tg2576 Green tea derived EGCG (50 mg/kg, oral in H2O) 6 months 8 months ↓ 50% in plaque load in hippocampus, cingulate and entorhinal cortex Improved performance in RAWM [199, 200]

Tg2576 EGCG (20 mg/kg,i.p.) 2 months 12 months ↓ 50 % in soluble Aβ40, 42
α-secretase cleavage by 40%
Not measured [200]

Tg2576 Luteolin (20 mg/kg, i.p.) 1 month 8 months ↓ GSK3 activity
↓ PS1 processing
Not measured [201]

Tg2576 Cabernet sauvignon wine (6% final ethanol concentration in H2O) 7 months 4 months ↓ Aβ40, 42 peptides in neocortex and hippocampus
↑ CTF cleaved fragment in neocortex
α-secretase activity
Improved performance in Barnes maze task [202]

3xTg Nicotine (0.025–0.6 mg/ml, in H2O) 5 months 1 months ↑ hippocampal Aβ42 AChRs
↑ tau phosphorylation in the hippocampus
↑ p38-MAP kinase
Not measured [209]

Tg2576 Nicotine (0.25–45 mg/kg, in H2O) 10 days 9 months ↓ soluble Aβ40, 42 in neocortex Not measured [207, 208]

Tg2576 Nicotine (25–35 mg/kg, in H2O) 5.5 months 9 months ↓ Aβ42 plaques in the neocortex, hippocampus, and olfactory bulb Not measured [207]

APPswe Curcumin (160–5000 ppm) 6 months 10 months ↓ oxidized proteins in neocortex and hippocampus
↓ soluble Aβ in neocortex and hippocampus
↓ total area and number of plaques
Not measured [214]

Tg2576 Ginko biloba (0.35 mg/kg, in H2O) 6 months 8 months No differences in plaque load Improved performance in MWM [216]

9. Strengths, Limitations, and Future Challenges of G×E Research in AD

This paper provides an overview of experimental mouse data on environmental exposures known to be associated with AD. In general, it may be concluded that many studies have shown effects of environmental manipulations on a wide variety of phenotypes in transgenic mouse models of AD.

It is challenging to evaluate the exact role of GxE in the field of preclinical AD research, due to several limitations. Numerous mouse models have been used on different genetic backgrounds and at multiple ages, applying various protocols of experimental exposures. This high level of variability makes it difficult to draw firm and general conclusions on any of the discussed exposures. In addition, the read-out parameters differed per group, with some focusing on Aβ pathology, synaptic integrity, or oxidative stress and others emphasizing behavioral effects. Another limitation is that most animal models used have focused on fAD mutations, while a GxE be more applicable in sporadic forms of AD.

The experimental design of most of the available studies often consisted of testing the effects of environmental exposures only in transgenic mice, without full comparison of the effects of those environmental exposures in wild type animals. In such an experimental setup, one can merely study disease acceleration or progression as a result of environmental exposure. Additionally, some environmental factors may have a differential effect in the initiation of the disease from their effect on disease progression. Evidently, this type of research can still provide us with insights on underlying biological mechanisms, but it cannot disentangle the synergistic participation of genes and environment in the induction of an AD phenotype.

In a clinical setting, however, GxE in AD etiology is particularly complex to decipher with standard epidemiological designs, particularly because the time-window between environmental exposures during life and the clinical phenotype of AD is very long. The high variability in environmental exposure across the life span also makes it challenging to capture this interaction.

Most AD research over the past decades has been Aβ-centered, yet recent clinical trials based on the amyloid cascade hypothesis have yielded controversial and sometimes disappointing results [219]. Similarly, the majority of animal models of AD have also grossly been focused on Aβ-enhancing mutations. Furthermore, outcome measures on the effects on environmental factors are often expressed in terms of soluble and insoluble Aβ and plaque load alterations. Because the extent of amyloid burden does not correlate well with AD symptomatology, one could argue that it would be advantageous to design future studies in a multidisciplinary manner encompassing a wide range of outcome measures such as behavioural phenotypes, biochemical, molecular, as well as neuropathological alterations and use similar outcome measures in the various studies conducted by different research groups so that results are more comparable.

Possible biological mechanisms that mediate the effects of environmental exposures and that could be the focus of further translational AD research are, among others, inflammation, oxidative stress, protein misfolding, glucose metabolism, and epigenetics [19, 220223]. The AD brain shows ample signs of ongoing inflammatory processes, such as the presence of proinflammatory cytokines and activated microglia surrounding amyloid plaques [224]. A large body of evidence has pointed towards a role of oxidative stress and oxidative damage in brain regions that are affected by AD [220]. In addition, changes in the epigenome have been implicated in the pathophysiology of AD that can be triggered by various environmental factors [19], while the exact role of misframed proteins, such as ubiquitin+1 that have been found to accumulate in AD, remains also to be fully elucidated [223, 225].

10. Concluding Remarks

It appears likely that a large part of, at least, sporadic cases can be connected with GxE, with the field being challenged to identify the most relevant GxE by for example conducting prospective clinical studies of subjects that will develop AD. Exposure to environmental risk factors during early life has been linked to other complex psychiatric phenotypes and disorders. For example, prenatal maternal and early life stress can be viewed as a risk factor for developing major depression and schizophrenia [226228]. Likewise, environmental exposures during early life may well impact on the risk of developing AD. For example, neonatal exposure to metal lead has been proposed as an early environmental trigger for AD-related pathology in rodents and macaque monkeys [109]. Findings like these have spurred researchers to formulate the “Latent Early Associated Regulation” (LEARn) theory of AD pathogenesis proposing that indeed early environmental exposures can change gene expression for long time periods and can induce pathology that only becomes apparent later in life, after subsequent trigger(s) [109, 229].

While numerous challenges lie ahead, it can be argued that it is timely to move attention of epidemiological as well as experimental animal research in the field of AD towards the synergistic approach of GxE research. In a preclinical setting, one could envision focusing more on the use of recently identified genetic variants of the newly found GWAS genes. Second, current studies can benefit from the further technological advances in transgenesis that enable time- and region-specific expression of transgenes, thereby allowing for the investigation of GxE during specific time windows in development and aging, as well as in specific brain regions. Third, it may be valuable to scrutinize the roles of genetic and environmental risk factors (and their interactions) of diseases that have been associated with the onset of AD, such as cardiovascular disease and diabetes mellitus type 2. The underlying mechanisms of these disorders, which likely involve GxE, could shed a new light on the etiology of AD.

To conclude, moving towards a GxE approach in both clinical and experimental animal studies seems promising in further elucidating the multifactorial etiology of AD, and in identifying modifiable factors that are of particular relevance for subgroups of AD patients. The further use and development of animal models combining genetic and environmental manipulations will be a driving force in elucidating the exact biological underpinnings of this detrimental disorder.

Acknowledgment

Funds were provided by the International Stichting Alzheimer Onderzoek (ISAO), Grant no. 09552 to B. P. F. Rutten and Grant no. 07551 to D. L. A. van den Hove; by a Marie Curie Host Fellowship Grant MC-EST 020589 EURON to L. Chouliaras; and by the National Institutes of Health (NIH) Grant no. AG05138 to P. R. Hof. L. Chouliaras and A. S. R. Sierksma are first joint to this study.

References

  • 1.Seshadri S, Wolf PA. Lifetime risk of stroke and dementia: current concepts, and estimates from the Framingham Study. Lancet Neurology. 2007;6(12):1106–1114. doi: 10.1016/S1474-4422(07)70291-0. [DOI] [PubMed] [Google Scholar]
  • 2.Querfurth HW, LaFerla FM. Alzheimer's disease. The New England Journal of Medicine. 2010;362(4):329–344. doi: 10.1056/NEJMra0909142. [DOI] [PubMed] [Google Scholar]
  • 3.Seshadri S, Fitzpatrick AL, Ikram MA, et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. Journal of the American Medical Association. 2010;303(18):1832–1840. doi: 10.1001/jama.2010.574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pedersen NL. Reaching the limits of genome-wide significance in Alzheimer disease: back to the environment. Journal of the American Medical Association. 2010;303(18):1864–1865. doi: 10.1001/jama.2010.609. [DOI] [PubMed] [Google Scholar]
  • 5.EU-GEI. Schizophrenia aetiology: do gene-environment interactions hold the key? Schizophrenia Research. 2008;102(1–3):21–26. doi: 10.1016/j.schres.2008.04.003. [DOI] [PubMed] [Google Scholar]
  • 6.Van Os J, Rutten BPF. Gene-environment-wide interaction studies in psychiatry. American Journal of Psychiatry. 2009;166(9):964–966. doi: 10.1176/appi.ajp.2008.09060904. [DOI] [PubMed] [Google Scholar]
  • 7.Van Os J, Rutten BPF, Poulton R. Gene-environment interactions in schizophrenia: review of epidemiological findings and future directions. Schizophrenia Bulletin. 2008;34(6):1066–1082. doi: 10.1093/schbul/sbn117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gatz M, Reynolds CA, Fratiglioni L, et al. Role of genes and environments for explaining Alzheimer disease. Archives of General Psychiatry. 2006;63(2):168–174. doi: 10.1001/archpsyc.63.2.168. [DOI] [PubMed] [Google Scholar]
  • 9.Räihä I, Kaprio J, Koskenvuo M, Rajala T, Sourander L. Alzheimer’s disease in Finnish twins. Lancet. 1996;347(9001):573–578. doi: 10.1016/s0140-6736(96)91272-6. [DOI] [PubMed] [Google Scholar]
  • 10.Räihä I, Kaprio J, Koskenvuo M, Rajala T, Sourander L. Environmental differences in twin pairs discordant for Alzheimer’s disease. Journal of Neurology, Neurosurgery and Psychiatry. 1998;65(5):785–787. doi: 10.1136/jnnp.65.5.785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Pedersen NL, Gatz M, Berg S, Johansson B. How heritable is Alzheimer’s disease late in life? Findings from Swedish twins. Annals of Neurology. 2004;55(2):180–185. doi: 10.1002/ana.10999. [DOI] [PubMed] [Google Scholar]
  • 12.Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nature Genetics. 2007;39(1):17–23. doi: 10.1038/ng1934. [DOI] [PubMed] [Google Scholar]
  • 13.Harold D, Abraham R, Hollingworth P, et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nature Genetics. 2009;41(10):1088–1093. doi: 10.1038/ng.440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lambert J-C, Heath S, Even G, et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nature Genetics. 2009;41(10):1094–1099. doi: 10.1038/ng.439. [DOI] [PubMed] [Google Scholar]
  • 15.Laumet G, Chouraki V, Grenier-Boley B, et al. Systematic analysis of candidate genes for Alzheimer's disease in a French, genome-wide association study. Journal of Alzheimer's Disease. 2010;20(4):1181–1188. doi: 10.3233/JAD-2010-100126. [DOI] [PubMed] [Google Scholar]
  • 16.Ganguli M, Kukull WA. Lost in translation: epidemiology, risk, and alzheimer disease. Archives of Neurology. 2010;67(1):107–111. doi: 10.1001/archneurol.2009.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Migliore L, Coppedè F. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutation Research. 2009;667(1-2):82–97. doi: 10.1016/j.mrfmmm.2008.10.011. [DOI] [PubMed] [Google Scholar]
  • 18.Qiu C, De Ronchi D, Fratiglioni L. The epidemiology of the dementias: an update. Current Opinion in Psychiatry. 2007;20(4):380–385. doi: 10.1097/YCO.0b013e32816ebc7b. [DOI] [PubMed] [Google Scholar]
  • 19.Chouliaras L, Rutten BPF, Kenis G, et al. Epigenetic regulation in the pathophysiology of Alzheimer's disease. Progress in Neurobiology. 2010;90(4):498–510. doi: 10.1016/j.pneurobio.2010.01.002. [DOI] [PubMed] [Google Scholar]
  • 20.Chandra V, Pandav R. Gene-environment interaction in Alzheimer’s disease: a potential role for cholesterol. Neuroepidemiology. 1998;17(5):225–232. doi: 10.1159/000026175. [DOI] [PubMed] [Google Scholar]
  • 21.Jarvik GP, Wijsman EM, Kukull WA, Schellenberg GD, Yu C, Larson EB. Interactions of apolipoprotein E genotype, total cholesterol level, age, and sex in prediction of Alzheimer’s disease: a case-control study. Neurology. 1995;45(6):1092–1096. doi: 10.1212/wnl.45.6.1092. [DOI] [PubMed] [Google Scholar]
  • 22.Elbaz A, Dufouil C, Alpérovitch A. Interaction between genes and environment in neurodegenerative diseases. Comptes Rendus Biologies. 2007;330(4):318–328. doi: 10.1016/j.crvi.2007.02.018. [DOI] [PubMed] [Google Scholar]
  • 23.Dufouil C, Tzourio C, Brayne C, Berr C, Amouyel P, Alpérovitch A. Influence of apolipoprotein E genotype on the risk of cognitive deterioration in moderate drinkers smokers. Epidemiology. 2000;11(3):280–284. doi: 10.1097/00001648-200005000-00009. [DOI] [PubMed] [Google Scholar]
  • 24.Håkansson K, Rovio S, Helkala EL, et al. Association between mid-life marital status and cognitive function in later life: population based cohort study. British Medical Journal. 2009;339:p. b2462. doi: 10.1136/bmj.b2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Basha MR, Murali M, Siddiqi HK, et al. Lead (Pb) exposure and its effect on APP proteolysis and Aβ aggregation. FASEB Journal. 2005;19(14):2083–2084. doi: 10.1096/fj.05-4375fje. [DOI] [PubMed] [Google Scholar]
  • 26.Oddo S, Caccamo A, Shepherd JD, et al. Triple-transgenic model of Alzheimer’s Disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron. 2003;39(3):409–421. doi: 10.1016/s0896-6273(03)00434-3. [DOI] [PubMed] [Google Scholar]
  • 27.Arendash GW, Sanchez-Ramos J, Mori T, et al. Electromagnetic field treatment protects against and reverses cognitive impairment in Alzheimer's disease mice. Journal of Alzheimer's Disease. 2010;19(1):191–210. doi: 10.3233/JAD-2010-1228. [DOI] [PubMed] [Google Scholar]
  • 28.Xu P-T, Schmechel D, Rothrock-Christian T, et al. Human apolipoprotein E2, E3, and E4 isoform-specific transgenic mice: human-like pattern of glial and neuronal immunoreactivity in central nervous system not observed in wild-type mice. Neurobiology of Disease. 1996;3(3):229–245. doi: 10.1006/nbdi.1996.0023. [DOI] [PubMed] [Google Scholar]
  • 29.Sturchler-Pierrat C, Abramowski D, Duke M, et al. Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(24):13287–13292. doi: 10.1073/pnas.94.24.13287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Blanchard V, Moussaoui S, Czech C, et al. Time sequence of maturation of dystrophic neurites associated with Aβ deposits in APP/PS1 transgenic mice. Experimental Neurology. 2003;184(1):247–263. doi: 10.1016/s0014-4886(03)00252-8. [DOI] [PubMed] [Google Scholar]
  • 31.Reaume AG, Howland DS, Trusko SP, et al. Enhanced amyloidogenic processing of the β-amyloid precursor protein in gene-targeted mice bearing the Swedish familial Alzheimer's disease mutations and a “humanized” Aβ sequence. Journal of Biological Chemistry. 1996;271(38):23380–23388. doi: 10.1074/jbc.271.38.23380. [DOI] [PubMed] [Google Scholar]
  • 32.Siman R, Reaume AG, Savage MJ, et al. Presenilin-1 P264L knock-in mutation: differential effects on Aβ production, amyloid deposition, and neuronal vulnerability. Journal of Neuroscience. 2000;20(23):8717–8726. doi: 10.1523/JNEUROSCI.20-23-08717.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Murphy MP, et al. Abeta solubility and deposition during AD progression and in APPxPS-1 knock-in mice. Neurobiology of Disease . 2007;27(3):301–311. doi: 10.1016/j.nbd.2007.06.002. [DOI] [PubMed] [Google Scholar]
  • 34.Mucke L, Masliah E, Yu G-Q, et al. High-level neuronal expression of Aβ(1-42) in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. Journal of Neuroscience. 2000;20(11):4050–4058. doi: 10.1523/JNEUROSCI.20-11-04050.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Savonenko A, Xu GM, Melnikova T, et al. Episodic-like memory deficits in the APPswe/PS1dE9 mouse model of Alzheimer's disease: relationships to β-amyloid deposition and neurotransmitter abnormalities. Neurobiology of Disease. 2005;18(3):602–617. doi: 10.1016/j.nbd.2004.10.022. [DOI] [PubMed] [Google Scholar]
  • 36.Li G, Jack CR, Yang X-F, Yang ES. Diet supplement CoQ10 delays brain atrophy in aged transgenic mice with mutations in the amyloid precursor protein: an in vivo volume MRI study. BioFactors. 2008;32(1–4):169–178. doi: 10.1002/biof.5520320120. [DOI] [PubMed] [Google Scholar]
  • 37.Yang X, Dai G, Li G, Yang ES. Coenzyme Q10 reduces β-amyloid plaque in an APP/PS1 transgenic mouse model of Alzheimer's disease. Journal of Molecular Neuroscience. 2010;41(1):110–113. doi: 10.1007/s12031-009-9297-1. [DOI] [PubMed] [Google Scholar]
  • 38.Holcomb L, Gordon MN, Mcgowan E, et al. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nature Medicine. 1998;4(1):97–100. doi: 10.1038/nm0198-097. [DOI] [PubMed] [Google Scholar]
  • 39.Li Q-X, Maynard C, Cappai R, et al. Intracellular accumulation of detergent-soluble amyloidogenic Aβ fragment of Alzheimer’s disease precursor protein in the hippocampus of aged transgenic mice. Journal of Neurochemistry. 1999;72(6):2479–2487. doi: 10.1046/j.1471-4159.1999.0722479.x. [DOI] [PubMed] [Google Scholar]
  • 40.Buxbaum JD, Christensen JL, Ruefli AA, Greengard P, Loring JF. Expression of APP in brains of transgenic mice containing the entire human APP gene. Biochemical and Biophysical Research Communications. 1993;197(2):639–645. doi: 10.1006/bbrc.1993.2527. [DOI] [PubMed] [Google Scholar]
  • 41.Duyckaerts C, Potier M-C, Delatour B. Alzheimer disease models and human neuropathology: similarities and differences. Acta Neuropathologica. 2008;115(1):5–38. doi: 10.1007/s00401-007-0312-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chui D-H, Tanahashi H, Ozawa K, et al. Transgenic mice with Alzheimer presenilin 1 mutations show accelerated neurodegeneration without amyloid plaque formation. Nature Medicine. 1999;5(5):560–564. doi: 10.1038/8438. [DOI] [PubMed] [Google Scholar]
  • 43.Howlett DR, Richardson JC, Austin A, et al. Cognitive correlates of Aβ deposition in male and female mice bearing amyloid precursor protein and presenilin-1 mutant transgenes. Brain Research. 2004;1017(1-2):130–136. doi: 10.1016/j.brainres.2004.05.029. [DOI] [PubMed] [Google Scholar]
  • 44.Gallant M, Rak M, Szeghalmi A, et al. Focally elevated creatine detected in amyloid precursor protein (APP) transgenic mice and alzheimer disease brain tissue. Journal of Biological Chemistry. 2006;281(1):5–8. doi: 10.1074/jbc.C500244200. [DOI] [PubMed] [Google Scholar]
  • 45.Hsiao K, Chapman P, Nilsen S, et al. Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice. Science. 1996;274(5284):99–102. doi: 10.1126/science.274.5284.99. [DOI] [PubMed] [Google Scholar]
  • 46.Chishti MA, Yang D-S, Janus C, et al. Early-onset amyloid deposition and cognitive deficits in transgenic mice expressing a double mutant form of amyloid precursor protein 695. Journal of Biological Chemistry. 2001;276(24):21562–21570. doi: 10.1074/jbc.M100710200. [DOI] [PubMed] [Google Scholar]
  • 47.Tanemura K, Murayama M, Akagi T, et al. Neurodegeneration with tau accumulation in a transgenic mouse expressing V337M human tau. Journal of Neuroscience. 2002;22(1):133–141. doi: 10.1523/JNEUROSCI.22-01-00133.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wilson RS, Arnold SE, Schneider JA, Kelly JF, Tang Y, Bennett DA. Chronic psychological distress and risk of Alzheimer’s disease in old age. Neuroepidemiology. 2006;27(3):143–153. doi: 10.1159/000095761. [DOI] [PubMed] [Google Scholar]
  • 49.Wilson RS, Evans DA, Bienias JL, Mendes De Leon CF, Schneider JA, Bennett DA. Proneness to psychological distress is associated with risk of Alzheimer’s disease. Neurology. 2003;61(11):1479–1485. doi: 10.1212/01.wnl.0000096167.56734.59. [DOI] [PubMed] [Google Scholar]
  • 50.Carroll BJ, Curtis GC. Neuroendocrine identification of depressed patients. Australian and New Zealand Journal of Psychiatry. 1976;10(1):13–20. doi: 10.3109/00048677609159480. [DOI] [PubMed] [Google Scholar]
  • 51.Csernansky JG, Dong H, Fagan AM, et al. Plasma cortisol and progression of dementia in subjects with Alzheimer-type dementia. American Journal of Psychiatry. 2006;163(12):2164–2169. doi: 10.1176/appi.ajp.163.12.2164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Weiner MF, Vobach S, Olsson K, Svetlik D, Risser RC. Cortisol secretion and Alzheimer’s disease progression. Biological Psychiatry. 1997;42(11):1030–1038. doi: 10.1016/s0006-3223(97)00165-0. [DOI] [PubMed] [Google Scholar]
  • 53.Joëls M, Karst H, Alfarez D, et al. Effects of chronic stress on structure and cell function in rat hippocampus and hypothalamus. Stress. 2004;7(4):221–231. doi: 10.1080/10253890500070005. [DOI] [PubMed] [Google Scholar]
  • 54.Magariños AM, McEwen BS, Flügge G, Fuchs E. Chronic psychosocial stress causes apical dendritic atrophy of hippocampal CA3 pyramidal neurons in subordinate tree shrews. Journal of Neuroscience. 1996;16(10):3534–3540. doi: 10.1523/JNEUROSCI.16-10-03534.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sapolsky RM. A mechanism for glucocorticoid toxicity in the hippocampus: increased neuronal vulnerability to metabolic insults. Journal of Neuroscience. 1985;5(5):1228–1232. doi: 10.1523/JNEUROSCI.05-05-01228.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sapolsky RM, Krey LC, McEwen BS. Prolonged glucocorticoid exposure reduces hippocampal neuron number: implications for aging. Journal of Neuroscience. 1985;5(5):1222–1227. doi: 10.1523/JNEUROSCI.05-05-01222.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Alfarez DN, Joëls M, Krugers HJ. Chronic unpredictable stress impairs long-term potentiation in rat hippocampal CA1 area and dentate gyrus in vitro. European Journal of Neuroscience. 2003;17(9):1928–1934. doi: 10.1046/j.1460-9568.2003.02622.x. [DOI] [PubMed] [Google Scholar]
  • 58.Gould E, McEwen BS, Tanapat P, Galea LAM, Fuchs E. Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. Journal of Neuroscience. 1997;17(7):2492–2498. doi: 10.1523/JNEUROSCI.17-07-02492.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sierksma ASR, van den Hove DLA, Steinbusch HWM, Prickaerts J. Major depression, cognitive dysfunction and Alzheimer’s disease: is there a link? European Journal of Pharmacology. 2010;626(1):72–82. doi: 10.1016/j.ejphar.2009.10.021. [DOI] [PubMed] [Google Scholar]
  • 60.Caraci F, Copani A, Nicoletti F, Drago F. Depression and Alzheimer’s disease: neurobiological links and common pharmacological targets. European Journal of Pharmacology. 2010;626(1):64–71. doi: 10.1016/j.ejphar.2009.10.022. [DOI] [PubMed] [Google Scholar]
  • 61.Rothman SM, Mattson MP. Adverse stress, hippocampal networks, and Alzheimer’s disease. NeuroMolecular Medicine. 2010;12(1):56–70. doi: 10.1007/s12017-009-8107-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Geerlings MI, Den Heijer T, Koudstaal PJ, Hofman A, Breteler MMB. History of depression, depressive symptoms, and medial temporal lobe atrophy and the risk of Alzheimer disease. Neurology. 2008;70(15):1258–1264. doi: 10.1212/01.wnl.0000308937.30473.d1. [DOI] [PubMed] [Google Scholar]
  • 63.Geerlings MI, Schoevers RA, Beekman ATF, et al. Depression and risk of cognitive decline and Alzheimer’s disease. Results of two prospective community-based studies in The Netherlands. British Journal of Psychiatry. 2000;176:568–575. doi: 10.1192/bjp.176.6.568. [DOI] [PubMed] [Google Scholar]
  • 64.Green RC, Cupples LA, Kurz A, et al. Depression as a risk factor for Alzheimer disease: the MIRAGE Study. Archives of Neurology. 2003;60(5):753–759. doi: 10.1001/archneur.60.5.753. [DOI] [PubMed] [Google Scholar]
  • 65.Grünblatt E, Monoranu CM, Apfelbacher M, et al. Tryptophan is a marker of human postmortem brain tissue quality. Journal of Neurochemistry. 2009;110(5):1400–1408. doi: 10.1111/j.1471-4159.2009.06233.x. [DOI] [PubMed] [Google Scholar]
  • 66.Devanand DP, Sano M, Tang M-X, et al. Depressed mood and the incidence of Alzheimer’s disease in the elderly living in the community. Archives of General Psychiatry. 1996;53(2):175–182. doi: 10.1001/archpsyc.1996.01830020093011. [DOI] [PubMed] [Google Scholar]
  • 67.Dotson VM, Beydoun MA, Zonderman AB. Recurrent depressive symptoms and the incidence of dementia and mild cognitive impairment. Neurology. 2010;75(1):27–34. doi: 10.1212/WNL.0b013e3181e62124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Saczynski JS, Beiser A, Seshadri S, Auerbach S, Wolf PA, Au R. Depressive symptoms and risk of dementia: the Framingham Heart Study. Neurology. 2010;75(1):35–41. doi: 10.1212/WNL.0b013e3181e62138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wilson RS, Hoganson GM, Rajan KB, Barnes LL, Mendes De Leon CF, Evans DA. Temporal course of depressive symptoms during the development of Alzheimer disease. Neurology. 2010;75(1):21–26. doi: 10.1212/WNL.0b013e3181e620c5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ownby DR, Johnson CC, Peterson EL. Incidence and prevalence of physician-diagnosed asthma in a suburban population of young adults. Annals of Allergy, Asthma and Immunology. 1996;77(4):304–308. doi: 10.1016/S1081-1206(10)63325-X. [DOI] [PubMed] [Google Scholar]
  • 71.Sun X, Chiu CC, Liebson E, et al. Depression and plasma amyloid β peptides in the elderly with and without the apolipoprotein E4 allele. Alzheimer Disease and Associated Disorders. 2009;23(3):238–244. doi: 10.1097/WAD.0b013e31819cb3ac. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.van Oijen M, Hofman A, Soares HD, Koudstaal PJ, Breteler MM. Plasma Aβ 1-40 and Aβ 1-42 and the risk of dementia: a prospective case-cohort study. Lancet Neurology. 2006;5(8):655–660. doi: 10.1016/S1474-4422(06)70501-4. [DOI] [PubMed] [Google Scholar]
  • 73.Lavretsky H, Siddarth P, Kepe V, et al. Depression and anxiety symptoms are associated with cerebral FDDNP-PET binding in middle-aged and older nondemented adults. American Journal of Geriatric Psychiatry. 2009;17(6):493–502. doi: 10.1097/jgp.0b013e3181953b82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Rapp MA, Schnaider-Beeri M, Grossman HT, et al. Increased hippocampal plaques and tangles in patients with Alzheimer disease with a lifetime history of major depression. Archives of General Psychiatry. 2006;63(2):161–167. doi: 10.1001/archpsyc.63.2.161. [DOI] [PubMed] [Google Scholar]
  • 75.Geerlings MI, Schmand B, Braam AW, Jonker C, Bouter LM, Van Tilburg W. Depressive symptoms and risk of Alzheimer’s disease in more highly educated older people. Journal of the American Geriatrics Society. 2000;48(9):1092–1097. doi: 10.1111/j.1532-5415.2000.tb04785.x. [DOI] [PubMed] [Google Scholar]
  • 76.Swaab DF, Bao A-M, Lucassen PJ. The stress system in the human brain in depression and neurodegeneration. Ageing Research Reviews. 2005;4(2):141–194. doi: 10.1016/j.arr.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 77.Dong H, Goico B, Martin M, Csernansky CA, Bertchume A, Csernansky JG. Modulation of hippocampal cell proliferation, memory, and amyloid plaque deposition in APPsw (Tg2576) mutant mice by isolation stress. Neuroscience. 2004;127(3):601–609. doi: 10.1016/j.neuroscience.2004.05.040. [DOI] [PubMed] [Google Scholar]
  • 78.Dong H, Yuede CM, Yoo H-S, et al. Corticosterone and related receptor expression are associated with increased β-amyloid plaques in isolated Tg2576 mice. Neuroscience. 2008;155(1):154–163. doi: 10.1016/j.neuroscience.2008.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kang J-E, Cirrito JR, Dong H, Csernansky JG, Holtzman DM. Acute stress increases interstitial fluid amyloid-β via corticotropin-releasing factor and neuronal activity. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(25):10673–10678. doi: 10.1073/pnas.0700148104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Pardon M-C, Sarmad S, Rattray I, et al. Repeated novel cage exposure-induced improvement of early Alzheimer’s-like cognitive and amyloid changes in TASTPM mice is unrelated to changes in brain endocannabinoids levels. Neurobiology of Aging. 2009;30(7):1099–1113. doi: 10.1016/j.neurobiolaging.2007.10.002. [DOI] [PubMed] [Google Scholar]
  • 81.Pedersen WA, Culmsee C, Ziegler D, Herman JP, Mattson MP. Aberrant stress response associated with severe hypoglycemia in a transgenic mouse model of Alzheimer’s disease. Journal of Molecular Neuroscience. 1999;13(1-2):159–165. doi: 10.1385/JMN:13:1-2:159. [DOI] [PubMed] [Google Scholar]
  • 82.Jeong YH, Park CH, Yoo J, et al. Chronic stress accelerates learning and memory impairments and increases amyloid deposition in APPV717I-CT100 transgenic mice, an Alzheimer’s disease model. FASEB Journal. 2006;20(6):729–731. doi: 10.1096/fj.05-4265fje. [DOI] [PubMed] [Google Scholar]
  • 83.Lee K-W, Kim J-B, Seo J-S, et al. Behavioral stress accelerates plaque pathogenesis in the brain of Tg2576 mice via generation of metabolic oxidative stress. Journal of Neurochemistry. 2009;108(1):165–175. doi: 10.1111/j.1471-4159.2008.05769.x. [DOI] [PubMed] [Google Scholar]
  • 84.Kunimoto S, Nakamura S, Wada K, Inoue T. Chronic stress-mutated presenilin 1 gene interaction perturbs neurogenesis and accelerates neurodegeneration. Experimental Neurology. 2009;221(1):175–185. doi: 10.1016/j.expneurol.2009.10.020. [DOI] [PubMed] [Google Scholar]
  • 85.Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM. Glucocorticoids increase amyloid-β and tau pathology in a mouse model of Alzheimer’s disease. Journal of Neuroscience. 2006;26(35):9047–9056. doi: 10.1523/JNEUROSCI.2797-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Boyle PA, Buchman AS, Barnes LL, Bennett DA. Effect of a purpose in life on risk of incident Alzheimer disease and mild cognitive impairment in community-dwelling older persons. Archives of General Psychiatry. 2010;67(3):304–310. doi: 10.1001/archgenpsychiatry.2009.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Laurin D, Verreault R, Lindsay J, MacPherson K, Rockwood K. Physical activity and risk of cognitive impairment and dementia in elderly persons. Archives of Neurology. 2001;58(3):498–504. doi: 10.1001/archneur.58.3.498. [DOI] [PubMed] [Google Scholar]
  • 88.Krech D, Rosenzweig MR, Bennett EL. Effects of environmental complexity and training on brain chemistry. Journal of Comparative and Physiological Psychology. 1960;53(6):509–519. doi: 10.1037/h0045402. [DOI] [PubMed] [Google Scholar]
  • 89.Nithianantharajah J, Hannan AJ. The neurobiology of brain and cognitive reserve: mental and physical activity as modulators of brain disorders. Progress in Neurobiology. 2009;89(4):369–382. doi: 10.1016/j.pneurobio.2009.10.001. [DOI] [PubMed] [Google Scholar]
  • 90.Adlard PA, Perreau VM, Pop V, Cotman CW. Voluntary exercise decreases amyloid load in a transgenic model of Alzheimer’s disease. Journal of Neuroscience. 2005;25(17):4217–4221. doi: 10.1523/JNEUROSCI.0496-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Arendash GW, Garcia MF, Costa DA, Cracchiolo JR, Wefes IM, Potter H. Environmental enrichment improves cognition in aged Alzheimer’s transgenic mice despite stable β-amyloid deposition. NeuroReport. 2004;15(11):1751–1754. doi: 10.1097/01.wnr.0000137183.68847.4e. [DOI] [PubMed] [Google Scholar]
  • 92.Herring A, Yasin H, Ambrée O, Sachser N, Paulus W, Keyvani K. Environmental enrichment counteracts Alzheimer’s neurovascular dysfunction in TgCRND8 mice. Brain Pathology. 2008;18(1):32–39. doi: 10.1111/j.1750-3639.2007.00094.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Herring A, Ambrée O, Tomm M, et al. Environmental enrichment enhances cellular plasticity in transgenic mice with Alzheimer-like pathology. Experimental Neurology. 2009;216(1):184–192. doi: 10.1016/j.expneurol.2008.11.027. [DOI] [PubMed] [Google Scholar]
  • 94.Görtz N, Lewejohann L, Tomm M, et al. Effects of environmental enrichment on exploration, anxiety, and memory in female TgCRND8 Alzheimer mice. Behavioural Brain Research. 2008;191(1):43–48. doi: 10.1016/j.bbr.2008.03.006. [DOI] [PubMed] [Google Scholar]
  • 95.Mirochnic S, Wolf S, Staufenbiel M, Kempermann G. Age effects on the regulation of adult hippocampal neurogenesis by physical activity and environmental enrichment in the APP23 mouse model of Alzheimer disease. Hippocampus. 2009;19(10):1008–1018. doi: 10.1002/hipo.20560. [DOI] [PubMed] [Google Scholar]
  • 96.Wolf SA, Kronenberg G, Lehmann K, et al. Cognitive and physical activity differently modulate disease progression in the amyloid precursor protein (APP)-23 model of Alzheimer’s disease. Biological Psychiatry. 2006;60(12):1314–1323. doi: 10.1016/j.biopsych.2006.04.004. [DOI] [PubMed] [Google Scholar]
  • 97.Cracchiolo JR, Mori T, Nazian SJ, Tan J, Potter H, Arendash GW. Enhanced cognitive activity-over and above social or physical activity-is required to protect Alzheimer’s mice against cognitive impairment, reduce Aβ deposition, and increase synaptic immunoreactivity. Neurobiology of Learning and Memory. 2007;88(3):277–294. doi: 10.1016/j.nlm.2007.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lazarov O, Robinson J, Tang Y-P, et al. Environmental enrichment reduces Aβ levels and amyloid deposition in transgenic mice. Cell. 2005;120(5):701–713. doi: 10.1016/j.cell.2005.01.015. [DOI] [PubMed] [Google Scholar]
  • 99.Costa DA, Cracchiolo JR, Bachstetter AD, et al. Enrichment improves cognition in AD mice by amyloid-related and unrelated mechanisms. Neurobiology of Aging. 2007;28(6):831–844. doi: 10.1016/j.neurobiolaging.2006.04.009. [DOI] [PubMed] [Google Scholar]
  • 100.Jankowsky JL, Xu G, Fromholt D, Gonzales V, Borchelt DR. Environmental enrichment exacerbates amyloid plaque formation in a transgenic mouse model of Alzheimer disease. Journal of Neuropathology and Experimental Neurology. 2003;62(12):1220–1227. doi: 10.1093/jnen/62.12.1220. [DOI] [PubMed] [Google Scholar]
  • 101.Jankowsky JL, Melnikova T, Fadale DJ, et al. Environmental enrichment mitigates cognitive deficits in a mouse model of Alzheimer’s disease. Journal of Neuroscience. 2005;25(21):5217–5224. doi: 10.1523/JNEUROSCI.5080-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Levi O, Jongen-Relo AL, Feldon J, Roses AD, Michaelson DM. ApoE4 impairs hippocampal plasticity isoform-specifically and blocks the environmental stimulation of synaptogenesis and memory. Neurobiology of Disease. 2003;13(3):273–282. doi: 10.1016/s0969-9961(03)00045-7. [DOI] [PubMed] [Google Scholar]
  • 103.Levi O, Dolev I, Belinson H, Michaelson DM. Intraneuronal amyloid-β plays a role in mediating the synergistic pathological effects of apoE4 and environmental stimulation. Journal of Neurochemistry. 2007;103(3):1031–1040. doi: 10.1111/j.1471-4159.2007.04810.x. [DOI] [PubMed] [Google Scholar]
  • 104.Nichol K, Deeny SP, Seif J, Camaclang K, Cotman CW. Exercise improves cognition and hippocampal plasticity in APOE epsilon4 mice. Alzheimer's and Dementia. 2009;5(4):287–294. doi: 10.1016/j.jalz.2009.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lewejohann L, Reefmann N, Widmann P, et al. Transgenic Alzheimer mice in a semi-naturalistic environment: more plaques, yet not compromised in daily life. Behavioural Brain Research. 2009;201(1):99–102. doi: 10.1016/j.bbr.2009.01.037. [DOI] [PubMed] [Google Scholar]
  • 106.French LR, Williams AN, Campbell RG. Lead toxicity. Minnesota Medicine. 1985;68(12):899–901. [PubMed] [Google Scholar]
  • 107.Gun RT, Korten AE, Jorm AF, et al. Occupational risk factors for Alzheimer disease: a case-control study. Alzheimer Disease and Associated Disorders. 1997;11(1):21–27. doi: 10.1097/00002093-199703000-00005. [DOI] [PubMed] [Google Scholar]
  • 108.Santibañez M, Bolumar F, García AM. Occupational risk factors in Alzheimer’s disease: a review assessing the quality of published epidemiological studies. Occupational and Environmental Medicine. 2007;64(11):723–732. doi: 10.1136/oem.2006.028209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Basha MR, Wei W, Bakheet SA, et al. The fetal basis of amyloidogenesis: exposure to lead and latent overexpression of amyloid precursor protein and β-amyloid in the aging brain. Journal of Neuroscience. 2005;25(4):823–829. doi: 10.1523/JNEUROSCI.4335-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wu J, Basha MR, Brock B, et al. Alzheimer’s Disease (AD)-like pathology in aged monkeys after infantile exposure to environmental metal lead (Pb): evidence for a developmental origin and environmental link for AD. Journal of Neuroscience. 2008;28(1):3–9. doi: 10.1523/JNEUROSCI.4405-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Flaten TP. Aluminium as a risk factor in Alzheimer’s disease, with emphasis on drinking water. Brain Research Bulletin. 2001;55(2):187–196. doi: 10.1016/s0361-9230(01)00459-2. [DOI] [PubMed] [Google Scholar]
  • 112.Rogers MAM, Simon DG. Aluminium intake and risk of Alzheimer's disease. Age and Ageing. 1999;28(2):205–209. doi: 10.1093/ageing/28.2.205. [DOI] [PubMed] [Google Scholar]
  • 113.Moore PB, Day JP, Taylor GA, Ferrier IN, Fifield LK, Edwardson JA. Absorption of aluminium-26 in Alzheimer’s disease, measured using accelerator mass spectrometry. Dementia and Geriatric Cognitive Disorders. 2000;11(2):66–69. doi: 10.1159/000017216. [DOI] [PubMed] [Google Scholar]
  • 114.Taylor GA, Ferrier IN, McLoughlin IJ, et al. Gastrointestinal absorption of aluminium in Alzheimer’s disease: response to aluminium citrate. Age and Ageing. 1992;21(2):81–90. doi: 10.1093/ageing/21.2.81. [DOI] [PubMed] [Google Scholar]
  • 115.Praticò D, Uryu K, Sung S, Tang S, Trojanowski JQ, Lee VM. Aluminum modulates brain amyloidosis through oxidative stress in APP transgenic mice. The FASEB Journal. 2002;16(9):1138–1140. doi: 10.1096/fj.02-0012fje. [DOI] [PubMed] [Google Scholar]
  • 116.Garcia T, Esparza J, Nogués MR, Romeu M, Domingo J, Gómez M. Oxidative stress status and RNA expression in hippocampus of an animal model of Alzheimer's disease after chronic exposure to aluminum. Hippocampus. 2010;20(1):218–225. doi: 10.1002/hipo.20612. [DOI] [PubMed] [Google Scholar]
  • 117.Ribes D, Colomina MT, Vicens P, Domingo JL. Effects of oral aluminum exposure on behavior and neurogenesis in a transgenic mouse model of Alzheimer’s disease. Experimental Neurology. 2008;214(2):293–300. doi: 10.1016/j.expneurol.2008.08.017. [DOI] [PubMed] [Google Scholar]
  • 118.Gómez M, Esparza JL, Cabré M, García T, Domingo JL. Aluminum exposure through the diet: metal levels in AβPP transgenic mice, a model for Alzheimer’s disease. Toxicology. 2008;249(2-3):214–219. doi: 10.1016/j.tox.2008.05.004. [DOI] [PubMed] [Google Scholar]
  • 119.Bush AI, Pettingell WH, Jr., Paradis MD, Tanzi RE. Modulation of Aβ adhesiveness and secretase site cleavage by zinc. Journal of Biological Chemistry. 1994;269(16):12152–12158. [PubMed] [Google Scholar]
  • 120.Miura T, Suzuki K, Kohata N, Takeuchi H. Metal binding modes of Alzheimer’s amyloid β-peptide in insoluble aggregates and soluble complexes. Biochemistry. 2000;39(23):7024–7031. doi: 10.1021/bi0002479. [DOI] [PubMed] [Google Scholar]
  • 121.Ricchelli F, Drago D, Filippi B, Tognon G, Zatta P. Aluminum-triggered structural modifications and aggregation of β-amyloids. Cellular and Molecular Life Sciences. 2005;62(15):1724–1733. doi: 10.1007/s00018-005-5141-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Dwyer BE, Zacharski LR, Balestra DJ, et al. Getting the iron out: phlebotomy for Alzheimer’s disease? Medical Hypotheses. 2009;72(5):504–509. doi: 10.1016/j.mehy.2008.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bartzokis G, Sultzer D, Cummings J, et al. In vivo evaluation of brain iron in Alzheimer disease using magnetic resonance imaging. Archives of General Psychiatry. 2000;57(1):47–53. doi: 10.1001/archpsyc.57.1.47. [DOI] [PubMed] [Google Scholar]
  • 124.Bartzokis G, Tishler TA, Shin I-S, Lu PH, Cummings JL. Brain ferritin iron as a risk factor for age at onset in neurodegenerative diseases. Annals of the New York Academy of Sciences. 2004;1012:224–236. doi: 10.1196/annals.1306.019. [DOI] [PubMed] [Google Scholar]
  • 125.Deibel MA, Ehmann WD, Markesbery WR. Copper, iron, and zinc imbalances in severely degenerated brain regions in Alzheimer’s disease: possible relation to oxidative stress. Journal of the Neurological Sciences. 1996;143(1-2):137–142. doi: 10.1016/s0022-510x(96)00203-1. [DOI] [PubMed] [Google Scholar]
  • 126.Lovell MA, Robertson JD, Teesdale WJ, Campbell JL, Markesbery WR. Copper, iron and zinc in Alzheimer’s disease senile plaques. Journal of the Neurological Sciences. 1998;158(1):47–52. doi: 10.1016/s0022-510x(98)00092-6. [DOI] [PubMed] [Google Scholar]
  • 127.Carlson ES, Magid R, Petryk A, Georgieff MK. Iron deficiency alters expression of genes implicated in Alzheimer disease pathogenesis. Brain Research. 2008;1237:75–83. doi: 10.1016/j.brainres.2008.07.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Jorgenson LA, Sun M, O’Connor M, Georgieff MK. Fetal iron deficiency disrupts the maturation of synaptic function and efficacy in area CA1 of the developing rat hippocampus. Hippocampus. 2005;15(8):1094–1102. doi: 10.1002/hipo.20128. [DOI] [PubMed] [Google Scholar]
  • 129.Jorgenson LA, Wobken JD, Georgieff MK. Perinatal iron deficiency alters apical dendritic growth in hippocampal CA1 pyramidal neurons. Developmental Neuroscience. 2003;25(6):412–420. doi: 10.1159/000075667. [DOI] [PubMed] [Google Scholar]
  • 130.Fernandez LL, Carmona M, Portero-Otin M, et al. Effects of increased iron intake during the neonatal period on the brain of adult AβPP/PS1 transgenic mice. Journal of Alzheimer's Disease. 2010;19(3):1069–1080. doi: 10.3233/JAD-2010-1304. [DOI] [PubMed] [Google Scholar]
  • 131.Moreira P, Pereira C, Santos MS, Oliveira C. Effect of zinc ions on the cytotoxicity induced by the amyloid β- peptide. Antioxidants and Redox Signaling. 2000;2(2):317–325. doi: 10.1089/ars.2000.2.2-317. [DOI] [PubMed] [Google Scholar]
  • 132.Carson JA, Turner AJ. β-amyloid catabolism: roles for neprilysin (NEP) and other metallopeptidases? Journal of Neurochemistry. 2002;81(1):1–8. doi: 10.1046/j.1471-4159.2002.00855.x. [DOI] [PubMed] [Google Scholar]
  • 133.Lovell MA, Xie C, Markesbery WR. Protection against amyloid beta peptide toxicity by zinc. Brain Research. 1999;823(1-2):88–95. doi: 10.1016/s0006-8993(99)01114-2. [DOI] [PubMed] [Google Scholar]
  • 134.Molina JA, Jiménez-Jiménez FJ, Aguilar MV, et al. Cerebrospinal fluid levels of transition metals in patients with Alzheimer’s disease. Journal of Neural Transmission. 1998;105(4-5):479–488. doi: 10.1007/s007020050071. [DOI] [PubMed] [Google Scholar]
  • 135.Yang D-S, McLaurin J, Qin K, Westaway D, Fraser PE. Examining the zinc binding site of the amyloid-β peptide. European Journal of Biochemistry. 2000;267(22):6692–6698. doi: 10.1046/j.1432-1327.2000.01767.x. [DOI] [PubMed] [Google Scholar]
  • 136.Stoltenberg M, Bush AI, Bach G, et al. Amyloid plaques arise from zinc-enriched cortical layers in APP/PS1 transgenic mice and are paradoxically enlarged with dietary zinc deficiency. Neuroscience. 2007;150(2):357–369. doi: 10.1016/j.neuroscience.2007.09.025. [DOI] [PubMed] [Google Scholar]
  • 137.Linkous DH, Adlard PA, Wanschura PB, Conko KM, Flinn JM. The effects of enhanced zinc on spatial memory and plaque formation in transgenic mice. Journal of Alzheimer's Disease. 2009;18(3):565–579. doi: 10.3233/JAD-2009-1162. [DOI] [PubMed] [Google Scholar]
  • 138.Maynard CJ, Cappai R, Volitakis I, et al. Chronic exposure to high levels of zinc or copper has little effect on brain metal homeostasis or Aβ accumulation in transgenic APP-C100 mice. Cellular and Molecular Neurobiology. 2009;29(5):757–767. doi: 10.1007/s10571-009-9401-7. [DOI] [PubMed] [Google Scholar]
  • 139.Lee J-Y, Cole TB, Palmiter RD, Suh SW, Koh J-Y. Contribution by synaptic zinc to the gender-disparate plaque formation in human Swedish mutant APP transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(11):7705–7710. doi: 10.1073/pnas.092034699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kessler H, Pajonk F-G, Bach D, et al. Effect of copper intake on CSF parameters in patients with mild Alzheimer’s disease: a pilot phase 2 clinical trial. Journal of Neural Transmission. 2008;115(12):1651–1659. doi: 10.1007/s00702-008-0136-2. [DOI] [PubMed] [Google Scholar]
  • 141.Sparks DL, Schreurs BG. Trace amounts of copper in water induce β-amyloid plaques and learning deficits in a rabbit model of Alzheimer's disease. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(19):11065–11069. doi: 10.1073/pnas.1832769100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Schäfer S, Pajonk F-G, Multhaup G, Bayer TA. Copper and clioquinol treatment in young APP transgenic and wild-type mice: effects on life expectancy, body weight, and metal-ion levels. Journal of Molecular Medicine. 2007;85(4):405–413. doi: 10.1007/s00109-006-0140-7. [DOI] [PubMed] [Google Scholar]
  • 143.Kitazawa M, Cheng D, Laferla FM. Chronic copper exposure exacerbates both amyloid and tau pathology and selectively dysregulates cdk5 in a mouse model of AD. Journal of Neurochemistry. 2009;108(6):1550–1560. doi: 10.1111/j.1471-4159.2009.05901.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Maynard CJ, Cappai R, Volitakis I, et al. Overexpression of Alzheimer’s disease amyloid-β opposes the age-dependent elevations of brain copper and iron. Journal of Biological Chemistry. 2002;277(47):44670–44676. doi: 10.1074/jbc.M204379200. [DOI] [PubMed] [Google Scholar]
  • 145.White AR, Reyes R, Mercer JFB, et al. Copper levels are increased in the cerebral cortex and liver of APP and APLP2 knockout mice. Brain Research. 1999;842(2):439–444. doi: 10.1016/s0006-8993(99)01861-2. [DOI] [PubMed] [Google Scholar]
  • 146.Mizoroki T, Meshitsuka S, Maeda S, Murayama M, Sahara N, Takashima A. Aluminum induces tau aggregation in vitro but not in vivo. Journal of Alzheimer’s Disease. 2007;11(4):419–427. doi: 10.3233/jad-2007-11401. [DOI] [PubMed] [Google Scholar]
  • 147.Johnson VE, Stewart W, Smith DH. Traumatic brain injury and amyloid-β pathology: a link to Alzheimer's disease? Nature Reviews Neuroscience. 2010;11(5):361–370. doi: 10.1038/nrn2808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Johnson VE, Stewart W, Graham DI, Stewart JE, Praestgaard AH, Smith DH. A neprilysin polymorphism and amyloid-β plaques after traumatic brain injury. Journal of Neurotrauma. 2009;26(8):1197–1202. doi: 10.1089/neu.2008.0843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Murai H, Pierce JES, Raghupathi R, et al. Twofold overexpression of human β-amyloid precursor proteins in transgenic mice does not affect the neuromotor, cognitive, or neurodegenerative sequelae following experimental brain injury. Journal of Comparative Neurology. 1998;392(4):428–438. doi: 10.1002/(sici)1096-9861(19980323)392:4<428::aid-cne2>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 150.Smith DH, Nakamura M, McIntosh TK, et al. Brain trauma induces massive hippocampal neuron death linked to a surge in β-amyloid levels in mice overexpressing mutant amyloid precursor protein. American Journal of Pathology. 1998;153(3):1005–1010. doi: 10.1016/s0002-9440(10)65643-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Abrahamson EE, Ikonomovic MD, Ciallella JR, et al. Caspase inhibition therapy abolishes brain trauma-induced increases in Aβ peptide: implications for clinical outcome. Experimental Neurology. 2006;197(2):437–450. doi: 10.1016/j.expneurol.2005.10.011. [DOI] [PubMed] [Google Scholar]
  • 152.Abrahamson EE, Ikonomovic MD, Edward Dixon C, DeKosky ST. Simvastatin therapy prevents brain trauma-induced increases in β-amyloid peptide levels. Annals of Neurology. 2009;66(3):407–414. doi: 10.1002/ana.21731. [DOI] [PubMed] [Google Scholar]
  • 153.Crawford FC, Wood M, Ferguson S, et al. Genomic analysis of response to traumatic brain injury in a mouse model of Alzheimer’s disease (APPsw) Brain Research. 2007;1185(1):45–58. doi: 10.1016/j.brainres.2007.09.042. [DOI] [PubMed] [Google Scholar]
  • 154.Uryu K, Laurer H, McIntosh T, et al. Repetitive mild brain trauma accelerates Aβ deposition, lipid peroxidation, and cognitive impairment in a transgenic mouse model of Alzheimer amyloidosis. Journal of Neuroscience. 2002;22(2):446–454. doi: 10.1523/JNEUROSCI.22-02-00446.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ferguson S, Mouzon B, Kayihan G, et al. Apolipoprotein E genotype and oxidative stress response to traumatic brain injury. Neuroscience. 2010;168(3):811–819. doi: 10.1016/j.neuroscience.2010.01.031. [DOI] [PubMed] [Google Scholar]
  • 156.Nakagawa Y, Nakamura M, McIntosh TK, et al. Traumatic brain injury in young, amyloid-β peptide overexpressing transgenic mice induces marked ipsilateral hippocampal atrophy and diminished deposition during aging. Journal of Comparative Neurology. 1999;411(3):390–398. [PubMed] [Google Scholar]
  • 157.Nakagawa Y, Reed L, Nakamura M, et al. Brain trauma in aged transgenic mice induces regression of established Aβ deposits. Experimental Neurology. 2000;163(1):244–252. doi: 10.1006/exnr.2000.7375. [DOI] [PubMed] [Google Scholar]
  • 158.Söderqvist F, Hardell L, Carlberg M, Mild KH. Radiofrequency fields, transthyretin, and alzheimer's disease. Journal of Alzheimer's Disease. 2010;20(2):599–606. doi: 10.3233/JAD-2010-1395. [DOI] [PubMed] [Google Scholar]
  • 159.Serot J-M, Christmann D, Dubost T, Couturier M. Cerebrospinal fluid transthyretin: aging and late onset Alzheimer’s disease. Journal of Neurology, Neurosurgery and Psychiatry. 1997;63(4):506–508. doi: 10.1136/jnnp.63.4.506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Galvin JE. Pass the grain; spare the brain. Neurology. 2007;69(11):1072–1073. doi: 10.1212/01.wnl.0000275542.03820.b1. [DOI] [PubMed] [Google Scholar]
  • 161.Scarmeas N, Stern Y, Mayeux R, Luchsinger JA. Mediterranean diet, alzheimer disease, and vascular mediation. Archives of Neurology. 2006;63(12):1709–1717. doi: 10.1001/archneur.63.12.noc60109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Scarmeas N, Stern Y, Mayeux R, Manly JJ, Schupf N, Luchsinger JA. Mediterranean diet and mild cognitive impairment. Archives of Neurology. 2009;66(2):216–225. doi: 10.1001/archneurol.2008.536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kivipelto M, Ngandu T, Fratiglioni L, et al. Obesity and vascular risk factors at midlife and the risk of dementia and Alzheimer disease. Archives of Neurology. 2005;62(10):1556–1560. doi: 10.1001/archneur.62.10.1556. [DOI] [PubMed] [Google Scholar]
  • 164.Luchsinger JA, Tang M-X, Shea S, Mayeux R. Caloric intake and the risk of Alzheimer disease. Archives of Neurology. 2002;59(8):1258–1263. doi: 10.1001/archneur.59.8.1258. [DOI] [PubMed] [Google Scholar]
  • 165.Pedrini S, Thomas C, Brautigam H, et al. Dietary composition modulates brain mass and solubilizable A levels in a mouse model of aggressive Alzheimer's amyloid pathology. Molecular Neurodegeneration. 2009;4(1):p. 40. doi: 10.1186/1750-1326-4-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ho L, Qin W, Pompl PN, et al. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. The FASEB Journal. 2004;18(7):902–904. doi: 10.1096/fj.03-0978fje. [DOI] [PubMed] [Google Scholar]
  • 167.Studzinski CM, Li F, Bruce-Keller AJ, et al. Effects of short-term Western diet on cerebral oxidative stress and diabetes related factors in APP x PS1 knock-in mice. Journal of Neurochemistry. 2009;108(4):860–866. doi: 10.1111/j.1471-4159.2008.05798.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Hooijmans CR, Rutters F, Dederen PJ, et al. Changes in cerebral blood volume and amyloid pathology in aged Alzheimer APP/PS1 mice on a docosahexaenoic acid (DHA) diet or cholesterol enriched Typical Western Diet (TWD) Neurobiology of Disease. 2007;28(1):16–29. doi: 10.1016/j.nbd.2007.06.007. [DOI] [PubMed] [Google Scholar]
  • 169.Julien C, Tremblay C, Phivilay A, et al. High-fat diet aggravates amyloid-beta and tau pathologies in the 3xTg-AD mouse model. Neurobiology of Aging. 2010;31(9):1516–1531. doi: 10.1016/j.neurobiolaging.2008.08.022. [DOI] [PubMed] [Google Scholar]
  • 170.Simons M, Keller P, Dichgans J, Schulz JB. Cholesterol and Alzheimer’s disease: is there a link? Neurology. 2001;57(6):1089–1093. doi: 10.1212/wnl.57.6.1089. [DOI] [PubMed] [Google Scholar]
  • 171.Notkola I-L, Sulkava R, Pekkanen J, et al. Serum total cholesterol, apolipoprotein E ε4 allele, and Alzheimer’s disease. Neuroepidemiology. 1998;17(1):14–20. doi: 10.1159/000026149. [DOI] [PubMed] [Google Scholar]
  • 172.Puglielli L, Tanzi RE, Kovacs DM. Alzheimer’s disease: the cholesterol connection. Nature Neuroscience. 2003;6(4):345–351. doi: 10.1038/nn0403-345. [DOI] [PubMed] [Google Scholar]
  • 173.Evans RM, Hui S, Perkins A, Lahiri DK, Poirier J, Farlow MR. Cholesterol and APOE genotype interact to influence Alzheimer disease progression. Neurology. 2004;62(10):1869–1871. doi: 10.1212/01.wnl.0000125323.15458.3f. [DOI] [PubMed] [Google Scholar]
  • 174.Kivipelto M, Helkala E-L, Laakso MP, et al. Apolipoprotein E ε4 allele, elevated midlife total cholesterol level, and high midlife systolic blood pressure are independent risk factors for late-life Alzheimer disease. Annals of Internal Medicine. 2002;137(3):149–155. doi: 10.7326/0003-4819-137-3-200208060-00006. [DOI] [PubMed] [Google Scholar]
  • 175.Refolo LM, Pappolla MA, Malester B, et al. Hypercholesterolemia accelerates the Alzheimer’s amyloid pathology in a transgenic mouse model. Neurobiology of Disease. 2000;7(4):321–331. doi: 10.1006/nbdi.2000.0304. [DOI] [PubMed] [Google Scholar]
  • 176.Refolo LM, Pappolla MA, LaFrancois J, et al. A cholesterol-lowering drug reduces β-amyloid pathology in a transgenic mouse model of Alzheimer's disease. Neurobiology of Disease. 2001;8(5):890–899. doi: 10.1006/nbdi.2001.0422. [DOI] [PubMed] [Google Scholar]
  • 177.Lim GP, Calon F, Morihara T, et al. A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model. Journal of Neuroscience. 2005;25(12):3032–3040. doi: 10.1523/JNEUROSCI.4225-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Oksman M, Iivonen H, Hogyes E, et al. Impact of different saturated fatty acid, polyunsaturated fatty acid and cholesterol containing diets on beta-amyloid accumulation in APP/PS1 transgenic mice. Neurobiology of Disease. 2006;23(3):563–572. doi: 10.1016/j.nbd.2006.04.013. [DOI] [PubMed] [Google Scholar]
  • 179.Green KN, Martinez-Coria H, Khashwji H, et al. Dietary docosahexaenoic acid and docosapentaenoic acid ameliorate amyloid-β and tau pathology via a mechanism involving presenilin 1 levels. Journal of Neuroscience. 2007;27(16):4385–4395. doi: 10.1523/JNEUROSCI.0055-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Perez SE, Berg BM, Moore KA, et al. DHA diet reduces AD pathology in young APPswe/PS1ΔE9 transgenic mice: possible gender effects. Journal of Neuroscience Research. 2010;88(5):1026–1040. doi: 10.1002/jnr.22266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Morris MC, Evans DA, Bienias JL, et al. Dietary fats and the risk of incident Alzheimer disease. Archives of Neurology. 2003;60(2):194–200. doi: 10.1001/archneur.60.2.194. [DOI] [PubMed] [Google Scholar]
  • 182.Morris MC, Evans DA, Bienias JL, et al. Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Archives of Neurology. 2003;60(7):940–946. doi: 10.1001/archneur.60.7.940. [DOI] [PubMed] [Google Scholar]
  • 183.Morris MC, Evans DA, Tangney CC, Bienias JL, Wilson RS. Fish consumption and cognitive decline with age in a large community study. Archives of Neurology. 2005;62(12):1849–1853. doi: 10.1001/archneur.62.12.noc50161. [DOI] [PubMed] [Google Scholar]
  • 184.Hooijmans CR, Van der Zee CEEM, Dederen PJ, et al. DHA and cholesterol containing diets influence Alzheimer-like pathology, cognition and cerebral vasculature in APPswe/PS1dE9 mice. Neurobiology of Disease. 2009;33(3):482–498. doi: 10.1016/j.nbd.2008.12.002. [DOI] [PubMed] [Google Scholar]
  • 185.Arendash GW, Jensen MT, Salem N, Jr., et al. A diet high in omega-3 fatty acids does not improve or protect cognitive performance in Alzheimer’s transgenic mice. Neuroscience. 2007;149(2):286–302. doi: 10.1016/j.neuroscience.2007.08.018. [DOI] [PubMed] [Google Scholar]
  • 186.Zhuo J-M, Praticò D. Acceleration of brain amyloidosis in an Alzheimer's disease mouse model by a folate, vitamin B6 and B12-deficient diet. Experimental Gerontology. 2010;45(3):195–201. doi: 10.1016/j.exger.2009.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Fuso A, Nicolia V, Pasqualato A, Fiorenza MT, Cavallaro RA, Scarpa S. Changes in Presenilin 1 gene methylation pattern in diet-induced B vitamin deficiency. doi: 10.1016/j.neurobiolaging.2009.02.013. Neurobiol. Aging 2010. In press. [DOI] [PubMed] [Google Scholar]
  • 188.Nicolia V, Fuso A, Cavallaro RA, Di Luzio A, Scarpa S. B vitamin deficiency promotes tau phosphorylation through regulation of GSK3β and PP2A. Journal of Alzheimer's Disease. 2010;19(3):895–907. doi: 10.3233/JAD-2010-1284. [DOI] [PubMed] [Google Scholar]
  • 189.Kruman II, Kumaravel TS, Lohani A, et al. Folic acid deficiency and homocysteine impair DNA repair in hippocampal neurons and sensitize them to amyloid toxicity in experimental models of Alzheimer’s disease. Journal of Neuroscience. 2002;22(5):1752–1762. doi: 10.1523/JNEUROSCI.22-05-01752.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Karuppagounder SS, Xu H, Shi Q, et al. Thiamine deficiency induces oxidative stress and exacerbates the plaque pathology in Alzheimer’s mouse model. Neurobiology of Aging. 2009;30(10):1587–1600. doi: 10.1016/j.neurobiolaging.2007.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Ding Y, Qiao A, Wang Z, et al. Retinoic acid attenuates β-amyloid deposition and rescues memory deficits in an Alzheimer’s disease transgenic mouse model. Journal of Neuroscience. 2008;28(45):11622–11634. doi: 10.1523/JNEUROSCI.3153-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Eskelinen MH, Kivipelto M. Caffeine as a protective factor in dementia and Alzheimer's disease. Journal of Alzheimer's Disease. 2010;20(supplement 1):S167–S174. doi: 10.3233/JAD-2010-1404. [DOI] [PubMed] [Google Scholar]
  • 193.Eskelinen MH, Ngandu T, Tuomilehto J, Soininen H, Kivipelto M. Midlife coffee and tea drinking and the risk of late-life dementia: a population-based CAIDE study. Journal of Alzheimer’s Disease. 2009;16(1):85–91. doi: 10.3233/JAD-2009-0920. [DOI] [PubMed] [Google Scholar]
  • 194.Santos C, et al. Caffeine intake is associated with a lower risk of cognitive decline: a cohort study from Portugal. Journal of Alzheimer's Disease. 2010;20(supplement 1):S175–S185. doi: 10.3233/JAD-2010-091303. [DOI] [PubMed] [Google Scholar]
  • 195.Santos C, et al. Caffeine intake and dementia: systematic review and meta-analysis. Journal of Alzheimer's Disease. 2010;20(supplement 1):S187–S204. doi: 10.3233/JAD-2010-091387. [DOI] [PubMed] [Google Scholar]
  • 196.Arendash GW, Mori T, Cao C, et al. Caffeine reverses cognitive impairment and decreases brain amyloid-β levels in aged alzheimer’s disease mice. Journal of Alzheimer’s Disease. 2009;17(3):661–680. doi: 10.3233/JAD-2009-1087. [DOI] [PubMed] [Google Scholar]
  • 197.Cao C, Cirrito JR, Lin X, et al. Caffeine suppresses amyloid-β levels in plasma and brain of alzheimer’s disease transgenic mice. Journal of Alzheimer’s Disease. 2009;17(3):681–697. doi: 10.3233/JAD-2009-1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Obregon DF, Rezai-Zadeh K, Bai Y, et al. ADAM10 activation is required for green tea (-)-epigallocatechin-3-gallate- induced α-secretase cleavage of amyloid precursor protein. Journal of Biological Chemistry. 2006;281(24):16419–16427. doi: 10.1074/jbc.M600617200. [DOI] [PubMed] [Google Scholar]
  • 199.Rezai-Zadeh K, Arendash GW, Hou H, et al. Green tea epigallocatechin-3-gallate (EGCG) reduces β-amyloid mediated cognitive impairment and modulates tau pathology in Alzheimer transgenic mice. Brain Research. 2008;1214:177–187. doi: 10.1016/j.brainres.2008.02.107. [DOI] [PubMed] [Google Scholar]
  • 200.Rezai-Zadeh K, Shytle D, Sun N, et al. Green tea epigallocatechin-3-gallate (EGCG) modulates amyloid precursor protein cleavage and reduces cerebral amyloidosis in Alzheimer transgenic mice. Journal of Neuroscience. 2005;25(38):8807–8814. doi: 10.1523/JNEUROSCI.1521-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Rezai-Zadeh K, Douglas Shytle R, Bai Y, et al. Flavonoid-mediated presenilin-1 phosphorylation reduces Alzheimer’s disease β-amyloid production. Journal of Cellular and Molecular Medicine. 2009;13(3):574–588. doi: 10.1111/j.1582-4934.2008.00344.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Wang J, Ho L, Zhao Z, et al. Moderate consumption of Cabernet Sauvignon attenuates Aβ neuropathology in a mouse model of Alzheimer's disease. FASEB Journal. 2006;20(13):2313–2320. doi: 10.1096/fj.06-6281com. [DOI] [PubMed] [Google Scholar]
  • 203.Ho L, Chen LH, Wang J, et al. Heterogeneity in red wine polyphenolic contents differentially influences Alzheimer’s disease-type neuropathology and cognitive deterioration. Journal of Alzheimer’s Disease. 2009;16(1):59–72. doi: 10.3233/JAD-2009-0916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Almeida OP, Garrido GJ, Lautenschlager NT, Hulse GK, Jamrozik K, Flicker L. Smoking is associated with reduced cortical regional gray matter density in brain regions associated with incipient alzheimer disease. American Journal of Geriatric Psychiatry. 2008;16(1):92–98. doi: 10.1097/JGP.0b013e318157cad2. [DOI] [PubMed] [Google Scholar]
  • 205.Almeida OP, Hulse GK, Lawrence D, Flicker L. Smoking as a risk factor for Alzheimer’s disease: contrasting evidence from a systematic review of case-control and cohort studies. Addiction. 2002;97(1):15–28. doi: 10.1046/j.1360-0443.2002.00016.x. [DOI] [PubMed] [Google Scholar]
  • 206.Cataldo JK, Prochaska JJ, Glantz SA. Cigarette smoking is a risk factor for Alzheimer's disease: an analysis controlling for tobacco industry affiliation. Journal of Alzheimer's Disease. 2010;19(2):465–480. doi: 10.3233/JAD-2010-1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Nordberg A, Hellström-Lindahl E, Lee M, et al. Chronic nicotine treatment reduces β-amyloidosis in the brain of a mouse model of Alzheimer's disease (APPsw) Journal of Neurochemistry. 2002;81(3):655–658. doi: 10.1046/j.1471-4159.2002.00874.x. [DOI] [PubMed] [Google Scholar]
  • 208.Hellström-Lindahl E, Court J, Keverne J, et al. Nicotine reduces Aβ in the brain and cerebral vessels of APPsw mice. European Journal of Neuroscience. 2004;19(10):2703–2710. doi: 10.1111/j.0953-816X.2004.03377.x. [DOI] [PubMed] [Google Scholar]
  • 209.Oddo S, Caccamo A, Green KN, et al. Chronic nicotine administration exacerbates tau pathology in a transgenic model of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(8):3046–3051. doi: 10.1073/pnas.0408500102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Patel NV, Gordon MN, Connor KE, et al. Caloric restriction attenuates Aβ-deposition in Alzheimer transgenic models. Neurobiology of Aging. 2005;26(7):995–1000. doi: 10.1016/j.neurobiolaging.2004.09.014. [DOI] [PubMed] [Google Scholar]
  • 211.Colman RJ, Anderson RM, Johnson SC, et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325(5937):201–204. doi: 10.1126/science.1173635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Halagappa VKM, Guo Z, Pearson M, et al. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease. Neurobiology of Disease. 2007;26(1):212–220. doi: 10.1016/j.nbd.2006.12.019. [DOI] [PubMed] [Google Scholar]
  • 213.Lim GP, Yang F, Chu T, et al. Ibuprofen effects on Alzheimer pathology and open field activity in APPsw transgenic mice. Neurobiology of Aging. 2001;22(6):983–991. doi: 10.1016/s0197-4580(01)00299-8. [DOI] [PubMed] [Google Scholar]
  • 214.Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. Journal of Neuroscience. 2001;21(21):8370–8377. doi: 10.1523/JNEUROSCI.21-21-08370.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Joseph JA, Denisova NA, Arendash G, et al. Blueberry supplementation enhances signaling and prevents behavioral deficits in an Alzheimer disease model. Nutritional Neuroscience. 2003;6(3):153–162. doi: 10.1080/1028415031000111282. [DOI] [PubMed] [Google Scholar]
  • 216.Stackman RW, Eckenstein F, Frei B, Kulhanek D, Nowlin J, Quinn JF. Prevention of age-related spatial memory deficits in a transgenic mouse model of Alzheimer’s disease by chronic Ginkgo biloba treatment. Experimental Neurology. 2003;184(1):510–520. doi: 10.1016/s0014-4886(03)00399-6. [DOI] [PubMed] [Google Scholar]
  • 217.Julien C, Tremblay C, Phivilay A, et al. High-fat diet aggravates amyloid-beta and tau pathologies in the 3xTg-AD mouse model. Neurobiology of Aging. 2010;31(9):1516–1531. doi: 10.1016/j.neurobiolaging.2008.08.022. [DOI] [PubMed] [Google Scholar]
  • 218.Arendash GW, Schleif W, Rezai-Zadeh K, et al. Caffeine protects Alzheimer’s mice against cognitive impairment and reduces brain β-amyloid production. Neuroscience. 2006;142(4):941–952. doi: 10.1016/j.neuroscience.2006.07.021. [DOI] [PubMed] [Google Scholar]
  • 219.Hardy J. The amyloid hypothesis for Alzheimer’s disease: a critical reappraisal. Journal of Neurochemistry. 2009;110(4):1129–1134. doi: 10.1111/j.1471-4159.2009.06181.x. [DOI] [PubMed] [Google Scholar]
  • 220.Rutten BPF, Steinbusch HWM, Korr H, Schmitz C. Antioxidants and Alzheimer’s disease: from bench to bedside (and back again) Current Opinion in Clinical Nutrition and Metabolic Care. 2002;5(6):645–651. doi: 10.1097/00075197-200211000-00006. [DOI] [PubMed] [Google Scholar]
  • 221.Rutten BPF, Van Der Kolk NM, Schafer S, et al. Age-related loss of synaptophysin immunoreactive presynaptic boutons within the hippocampus of APP751SL, PS1M146L, and APP751 SL/PS1M146L transgenic mice. American Journal of Pathology. 2005;167(1):161–173. doi: 10.1016/S0002-9440(10)62963-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Schmitz C, Rutten BPF, Pielen A, et al. Hippocampal neuron loss exceeds amyloid plaque load in a transgenic mouse model of Alzheimer’s disease. American Journal of Pathology. 2004;164(4):1495–1502. doi: 10.1016/S0002-9440(10)63235-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Dennissen FJA, Kholod N, Steinbusch HWM, Van Leeuwen FW. Misframed proteins and neurodegeneration: a novel view on Alzheimer's and Parkinson's diseases. Neurodegenerative Diseases. 2010;7(1–3):76–79. doi: 10.1159/000285510. [DOI] [PubMed] [Google Scholar]
  • 224.McNaull BBA, Todd S, McGuinness B, Passmore AP. Inflammation and anti-inflammatory strategies for Alzheimer's disease—a mini-review. Gerontology. 2010;56(1):3–14. doi: 10.1159/000237873. [DOI] [PubMed] [Google Scholar]
  • 225.Van Leeuwen FW, De Kleijn DPV, Van Den Hurk HH, et al. Frameshift mutants of β amyloid precursor protein and ubiquitin-B in Alzheimer’s and Down patients. Science. 1998;279(5348):242–247. doi: 10.1126/science.279.5348.242. [DOI] [PubMed] [Google Scholar]
  • 226.Brown ES, Fulton MK, Wilkeson A, Petty F. The psychiatric sequelae of civilian trauma. Comprehensive Psychiatry. 2000;41(1):19–23. doi: 10.1016/s0010-440x(00)90126-3. [DOI] [PubMed] [Google Scholar]
  • 227.Rutten BP, Mill J. Epigenetic mediation of environmental influences in major psychotic disorders. Schizophrenia Bulletin. 2009;35(6):1045–1056. doi: 10.1093/schbul/sbp104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Van Os J, Selten J-P. Prenatal exposure to maternal stress and subsequent schizophrenia. The May 1940 invasion of The Netherlands. British Journal of Psychiatry. 1998;172:324–326. doi: 10.1192/bjp.172.4.324. [DOI] [PubMed] [Google Scholar]
  • 229.Lahiri DK, Maloney B, Zawia NH. The LEARn model: an epigenetic explanation for idiopathic neurobiological diseases. Molecular Psychiatry. 2009;14(11):992–1003. doi: 10.1038/mp.2009.82. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from International Journal of Alzheimer's Disease are provided here courtesy of Wiley

RESOURCES